CN116745292A - Substituted pyridotriazine compounds and uses thereof - Google Patents

Substituted pyridotriazine compounds and uses thereof Download PDF

Info

Publication number
CN116745292A
CN116745292A CN202280010649.4A CN202280010649A CN116745292A CN 116745292 A CN116745292 A CN 116745292A CN 202280010649 A CN202280010649 A CN 202280010649A CN 116745292 A CN116745292 A CN 116745292A
Authority
CN
China
Prior art keywords
formula
compound
pharmaceutically acceptable
acceptable salt
alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280010649.4A
Other languages
Chinese (zh)
Inventor
储航
A·Z·冈萨雷斯·布恩罗斯特罗
郭洪燕
韩晓春
A·E·赫特利
姜岚
李加尧
D·W·林
M·L·米歇尔
D·纳杜撒比
G·M·施瓦兹瓦尔德
S·M·谢夫奇克
M·J·范巴根
吴巧银
杨红
J·R·张
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Gilead Sciences Inc
Original Assignee
Gilead Sciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gilead Sciences Inc filed Critical Gilead Sciences Inc
Priority claimed from PCT/US2022/012773 external-priority patent/WO2022159387A1/en
Publication of CN116745292A publication Critical patent/CN116745292A/en
Pending legal-status Critical Current

Links

Abstract

The present disclosure relates generally to certain tricyclic compounds, pharmaceutical compositions comprising the compounds, and methods of preparing the compounds and pharmaceutical compositions. The compounds of the present disclosure are useful for treating or preventing Human Immunodeficiency Virus (HIV) infection.

Description

Substituted pyridotriazine compounds and uses thereof
Cross Reference to Related Applications
The present application claims priority from U.S. provisional patent application number 63/139,237, filed on day 19 of 1 month 2021, and U.S. provisional patent application number 63/190,461, filed on day 19 of 5 month 2021, each of which is incorporated herein in its entirety for all purposes.
Technical Field
The present disclosure relates generally to certain 2, 3-dihydro-1H-pyrido [2,1-f ] [1,2,4] triazine compounds, pharmaceutical compositions comprising the compounds, and methods of making and using the compounds and pharmaceutical compositions.
Background
Human immunodeficiency virus infection and related diseases are a major public health problem worldwide. Human immunodeficiency virus encodes three enzymes required for viral replication: reverse transcriptase, protease and integrase. Although drugs targeting reverse transcriptase and protease have been widely used and have been shown to be effective, particularly when used in combination, the development of toxic and drug-resistant strains may limit their usefulness (Palella et al, n.engl.j. Med. (1998) 338:853-860; richman, d.d. nature (2001) 410:995-1001). Thus, new agents that inhibit HIV replication are needed.
One goal of antiretroviral therapy is to achieve viral suppression in HIV-infected patients. Current therapeutic guidelines issued by the U.S. health and public service department specify that achieving viral suppression requires the use of combination therapy, i.e., several drugs in at least two or more drug categories (Panel on Antiretroviral Guidelines for Adults and advertisements, guides for the Use of Antiretroviral Agents in Adults and Adolescents Living with hiv. Components of Health and Human services, website: https:// files, aids info. Nih. Gov/content files/lvguide/addda doscore gl. Pdf.2020, 20-day access for 2 months). In addition, decisions regarding treatment of HIV-infected patients are complex when the patient needs to treat other medical conditions. Because the standard of care requires the use of a number of different drugs to inhibit HIV, as well as to treat other conditions that a patient may be experiencing, the possibility of drug interactions is the standard of choice for drug regimens. Thus, there is a need for antiretroviral therapy with reduced potential for drug interactions.
In addition, HIV virus is known to mutate in infected subjects (Tang et al Drugs (2012) 72 (9) e1-e 25). Because of the propensity of the HIV virus to mutate, there is a need for anti-HIV drugs that are effective against a range of known HIV variants (Hurt et al HIV/AIDS CID (2014) 58, 423-431).
For some patients, such as patients with difficulty or limited in achieving healthcare, adherence to a daily oral treatment or prevention regimen can be challenging. Drugs that provide advantageous pharmaceutical properties (e.g., improved potency, long-acting pharmacokinetics, low solubility, low clearance, and/or other properties) may reduce the frequency of administration and provide better patient compliance. Such improvements in turn may optimize drug exposure and limit the emergence of drug resistance.
Disclosure of Invention
In some embodiments, disclosed herein are compounds of formula I:
or a pharmaceutically acceptable salt thereof, wherein
Ar is C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S; wherein said C 6 -C 10 Aryl or six to ten membered heteroaryl optionally being 1 to 4 independently selected from halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl and C 1 -C 6 Substituent substitution of the group consisting of alkoxy;
R 1 h, C of a shape of H, C 1 -C 3 Alkyl or phenyl;
R 2 is H or C 1 -C 3 An alkyl group;
R 3 is H or C 1 -C 3 An alkyl group;
R 4 and R is 5 Each independently is H, halogen, cyano, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S; wherein said C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or six-to ten-membered heteroaryl groups optionally substituted with one, two or moreThree are independently selected from halogen, C 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; or alternatively
R 4 And R is 5 Taken together form a 3-6 membered carbocyclic ring or a 4-6 membered heterocyclic ring containing one heteroatom selected from N, O and S;
R 6 is H, halogen or C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group;
R 7 is H, halogen or C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group;
R 8A and R is 8B H, C independently 1 -C 3 Alkyl or benzyl; and is also provided with
-X-Y-is- (CR) 13A R 13B ) p -CR 9 =CR 10 -, -or- (CR) 13A R 13B ) q -CR 11A R 11B -CR 12A R 12B -; wherein the method comprises the steps of
R 9 Is H, halogen or C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or C 1 -C 6 An alkoxy group;
R 10 is H, halogen or C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or C 1 -C 6 An alkoxy group; or alternatively
R 9 And R is 10 Together with the carbon to which they are attached, form a phenyl group or a 5-6 membered heteroaryl ring containing 1, 2 or 3 heteroatoms independently selected from N, O and S; wherein said phenyl or said 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 Substituent substitution of the group consisting of alkoxy; and is also provided with
R 11A 、R 11B 、R 12A 、R 12B 、R 13A And R is 13B Each independently is H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 An alkoxy group; or C 1 -C 6 A haloalkyl group; or alternatively
R 11A 、R 12A 、R 13A And R is 13B Each independently is H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group; and R is 11B And R is 12B Together with the carbon to which they are attached, form a 3-6 membered carbocyclic ring. Wherein the 3-6 membered carbocycle is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 Substituent substitution of the group consisting of alkoxy;
p is 0 or 1;
q is 0 or 1;
wherein when-X-Y is- (CR) 13A R 13B ) q -CR 11A R 11B -CR 12A R 12B -when, then:
(i)R 4 is halogen, cyano, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S; wherein said C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or six-to ten-membered heteroaryl optionally being independently selected from halogen, C 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; and R is 5 Is H, halogen, cyano, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S; wherein said C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or six-to ten-membered heteroaryl optionally being independently selected from halogen, C 1 -C 3 Alkoxy or C 1 -C 3 Group extraction of haloalkoxy groupsSubstitution; or alternatively
(ii)R 4 And R is 5 Are linked together to form a 3-6 membered carbocyclic ring or a 4-6 membered heterocyclic ring having one heteroatom; or alternatively
(iii)R 8A Is C 1 -C 3 Alkyl or benzyl; or alternatively
(iv)R 6 Is halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group.
In some embodiments, the present disclosure provides pharmaceutical compositions comprising a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
In some embodiments, the present disclosure provides a kit comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and instructions for use.
In some embodiments, the present disclosure provides a method of treating HIV infection in a human having or at risk of having HIV infection, the method comprising administering to the human a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
In some embodiments, the present disclosure provides the use of a compound of formula I, a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for treating HIV infection in a human suffering from or at risk of suffering from HIV infection.
In some embodiments, the present disclosure provides a compound of formula I, a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one thereof for use in medical therapy.
In some embodiments, the present disclosure provides a compound of formula I, a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one thereof for use in treating HIV infection.
In some embodiments, the present disclosure provides the use of a compound of formula I, a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for treating HIV infection in a human having or at risk of having HIV infection.
Detailed Description
In the following description, certain specific details are set forth in order to provide a thorough understanding of the various embodiments disclosed herein. However, it will be understood by those skilled in the art that the embodiments disclosed herein may be practiced without these details. The following description of several embodiments is provided with the understanding that the present disclosure is to be considered as an exemplification of the claimed subject matter and is not intended to limit the appended claims to the specific embodiments illustrated. Headings used throughout this disclosure are provided for convenience only and should not be construed as limiting the claims in any way. The embodiments illustrated under any heading may be combined with the embodiments illustrated under any other heading.
I. Definition of the definition
Throughout this disclosure and the claims, unless the context requires otherwise, the words "comprise" and variations such as "comprises" and "comprising" are to be construed in an open, inclusive sense, i.e. as "including but not limited to".
Reference throughout this specification to "one embodiment" or "an embodiment" means that a particular feature, structure, or characteristic described in connection with the embodiment is included in at least one embodiment disclosed herein. Thus, the appearances of the phrase "in one embodiment" or "in an embodiment" appearing in various places throughout the specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments.
"amino" means-NH 2 A group.
"hydroxy" refers to an-OH group.
"oxo" refers to an =o substituent.
Prefix such as "C u-v Or (C) u -C v ) The latter groups are represented as having u to v carbon atoms. For example, "C 1-6 Alkyl "or" C 1 -C 6 Alkyl "indicates that the alkyl group has 1 to 6 carbon atoms.
"alkyl" means a straight or branched hydrocarbon group consisting of carbon and hydrogen atoms, which is saturated and has one to twelve carbon atoms (C 1-12 Alkyl groups), in certain embodiments having one to eight carbon atoms (C 1-8 Alkyl) or one to six carbon atoms (C 1-6 Alkyl) or one to four carbon atoms (C 1-4 Alkyl) and is attached to the remainder of the molecule by a single bond, such as methyl, ethyl, n-propyl, 1-methylethyl (isopropyl), n-butyl, 1-methylpropyl (sec-butyl), 2-methylpropyl (isobutyl), 1-dimethylethyl (tert-butyl), n-pentyl, hexyl, 3-methylhexyl, 2-methylhexyl, and the like.
"alkylene" refers to a saturated, branched or straight chain or cyclic hydrocarbon group having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of the parent alkane. For example, the alkylene group may have 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. Typical alkylene groups include, but are not limited to, methylene (-CH) 2 (-), 1-ethyl (-CH (CH) 3 ) (-), 1, 2-ethyl (-CH) 2 CH 2 (-), 1-propyl (-CH (CH) 2 CH 3 ) (-), 1, 2-propyl (-CH) 2 CH(CH 3 ) (-), 1, 3-propyl (-CH) 2 CH 2 CH 2 (-), 1, 4-butyl (-CH) 2 CH 2 CH 2 CH 2 (-), etc.
"aryl" or "aromatic ring" refers to an aromatic carbocyclic group having a single ring (e.g., a single ring) or containing multiple rings (e.g., bicyclic or tricyclic) of a fused system. As used herein, aryl groups have 6 to 20 ring carbon atoms (i.e., C 6-20 Aryl), 6 to 12 carbon ring atoms (i.e. C 6-12 Aryl) or 6 to 10 carbon ring atoms (i.e. C 6-10 Aryl). Examples of aryl groups include, but are not limited to, phenyl, naphthyl, fluorenyl, and anthracyl. However, aryl does not encompass or overlap in any way with heteroaryl as defined below.
"cyano" or "carbonitrile" refers to the group-CN.
"cycloalkyl" or "carbocycle" refers to a saturated or carbocyclic ring having a single ring or multiple rings comprising fused, bridged and spiro ring systemsPartially saturated cyclic alkyl groups. The term "cycloalkyl" includes cycloalkenyl groups (i.e., cyclic groups having at least one double bond). As used herein, cycloalkyl groups have 3 to 20 ring carbon atoms (i.e., C 3-20 Cycloalkyl), 3 to 12 ring carbon atoms (i.e., C 3-12 Cycloalkyl), 3 to 10 ring carbon atoms (i.e., C 3-10 Cycloalkyl), 3 to 8 ring carbon atoms (i.e., C 3-8 Cycloalkyl) or 3 to 6 ring carbon atoms (i.e., C 3-6 Cycloalkyl). Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. "halocycloalkyl" refers to cycloalkyl substituted with one or more halogens.
"halo" or "halogen" refers to bromine, chlorine, fluorine, or iodine.
"haloalkyl" refers to an alkyl group as defined above substituted with one or more halo groups as defined above, such as trifluoromethyl, difluoromethyl, trichloromethyl, 2-trifluoroethyl, 1, 2-difluoroethyl, 3-bromo-2-fluoropropyl, 1, 2-dibromoethyl and the like.
"heteroaryl" or "heteroaromatic ring" refers to an aromatic group having a single ring, multiple rings, or multiple fused rings, wherein one or more ring heteroatoms are independently selected from nitrogen, oxygen, and sulfur. As used herein, heteroaryl includes 5 to 20 ring atoms (5 to 20 membered heteroaryl ring), 5 to 12 ring atoms (5 to 12 membered heteroaryl ring), 5 to 10 ring atoms (5 to 10 membered heteroaryl ring), or 5 to 6 ring atoms (5 to 6 membered heteroaryl ring); and 1 to 5 ring heteroatoms, 1 to 4 ring heteroatoms, 1 to 3 ring heteroatoms, 1 to 2 ring heteroatoms, or 1 ring heteroatom independently selected from nitrogen, oxygen, and sulfur. Examples of heteroaryl groups include pyrimidinyl, purinyl, pyridinyl, pyridazinyl, benzothiazolyl, and pyrazolyl. Heteroaryl does not encompass or overlap with aryl as defined above.
"heterocyclyl" or "heterocycle" refers to a non-aromatic group or ring having three to fifteen atoms, where one to six atoms are heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur and are attached to the remainder of the molecule by single bonds. In certain embodiments, a "heterocyclyl" has three to ten atoms, one to four of which are heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur, or three to seven atoms, one to two of which are heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur. The nitrogen, carbon or sulfur atoms in the heterocyclyl group may optionally be oxidized; the nitrogen atom may optionally be quaternized. As used herein, unless otherwise indicated, "heterocyclyl" or "heterocycle" refers to a saturated ring, e.g., in some embodiments, "heterocyclyl" or "heterocycle" refers to a saturated or partially saturated ring where specified. Examples of such heterocyclic groups include, but are not limited to, dioxolanyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidinyl, pyrrolidinyl, pyrazolidinyl, thiazolidinyl, tetrahydrofuranyl, trithioheterocycloalkyl, tetrahydropyranyl, thiomorpholinyl, 1-oxo-thiomorpholinyl, and 1, 1-dioxo-thiomorpholinyl.
Embodiments disclosed herein are also intended to encompass all pharmaceutically acceptable compounds of formula I isotopically labeled by having one or more atoms replaced by atoms having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine, chlorine, and iodine, respectively, such as 2 H、 3 H、 11 C、 13 C、 14 C、 13 N、 15 N、 15 O、 17 O、 18 O、 31 P、 32 P、 35 S、 18 F、 36 Cl、 123 I and 125 I. in certain embodiments, these radiolabeled compounds may be used to help determine or measure the effectiveness of a compound by characterizing, for example, the site or pattern of action or binding affinity to a pharmacologically important site of action. Certain isotopically-labeled compounds of formula I, ia, ib, II, IIa, IIb, III, IIIa, IIIb, IV, IVa, IVb, V, va or Vb (e.g., those into which a radioisotope is incorporated) are useful in pharmaceutical and/or substrate tissue distribution studies. Radioisotope tritium (i.e 3 H) And carbon-14 (i.e 14 C) Are particularly suitable for this purpose because they are easy to incorporate and the detection means are convenient.
In certain embodiments, the heavy isotopes such as deuterium (i.e 2 H) Substitution may provide certain therapeutic advantages due to higher metabolic stability. For example, in vivo half-life may be increased or dosage requirements may be reduced. Thus, in some cases, heavier isotopes may be preferred.
By positron-emitting isotopes such as 11 C、 18 F、 15 O and 13 n-substitution can be used in Positron Emission Tomography (PET) studies to examine substrate receptor occupancy. Isotopically-labeled compounds of formula I, ia, ib, II, IIa, IIb, III, IIIa, IIIb, IV, IVa, IVb, V, va or Vb can be prepared by techniques known to those skilled in the art or by processes analogous to those described in the examples set forth below using an appropriate isotopically-labeled reagent in place of the previously employed non-labeled reagent.
The methods, compositions, kits and articles of manufacture provided herein use or include a compound (e.g., a compound of formula I, ia, ib, II, IIa, IIb, III, IIIa, IIIb, IV, IVa, IVb, V, va or Vb) or a pharmaceutically acceptable salt thereof, wherein 1 to n hydrogen atoms attached to a carbon atom can be replaced with a deuterium atom or D, where n is the number of hydrogen atoms in the molecule. As known in the art, deuterium atoms are nonradioactive isotopes of hydrogen atoms. Such compounds will increase resistance to metabolism and thus may be useful to increase the half-life of the compound or pharmaceutically acceptable salt thereof when administered to a mammal. See, e.g., foster, "Deuterium Isotope Effects in Studies of Drug Metabolism", trends Pharmacol. Sci.,5 (12): 524-527 (1984). Such compounds may be synthesized by means known in the art, for example, by employing starting materials in which one or more hydrogen atoms have been replaced by deuterium.
Embodiments disclosed herein are also intended to encompass in vivo metabolites of the disclosed compounds. Such products may result, for example, from oxidation, reduction, hydrolysis, amidation, esterification, etc., of the applied compounds, mainly due to enzymatic processes. Thus, embodiments disclosed herein include compounds produced by a process comprising administering a compound according to embodiments disclosed herein to a mammal for a period of time sufficient to produce a metabolite thereof. Such products are typically identified by administering a radiolabeled compound according to embodiments disclosed herein to an animal (such as a rat, mouse, guinea pig, monkey, or human) at a detectable dose, allowing sufficient time for metabolism to occur and isolation of their conversion products from urine, blood, or other biological samples.
"mammal" includes humans and domestic animals such as laboratory animals and domestic pets (e.g., cats, dogs, pigs, cattle, sheep, goats, horses, rabbits) and non-domestic animals such as wild animals and the like.
"optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. For example, "optionally substituted heterocyclyl" means that the heterocyclyl may or may not be substituted, and the description includes both substituted heterocyclyl and unsubstituted heterocyclyl.
"pharmaceutically acceptable excipients" include, but are not limited to, any adjuvants, carriers, excipients, glidants, sweeteners, diluents, preservatives, dyes/colorants, flavor enhancers, surfactants, wetting agents, dispersants, suspending agents, stabilizers, isotonic agents, solvents, emulsifying agents or other pharmacologically inactive substances formulated in combination with the pharmacologically active ingredient of a pharmaceutical composition and compatible with the other ingredients of the formulation and suitable for use in humans or livestock without undue toxicity, irritation, allergic response, and the like.
Examples of "pharmaceutically acceptable salts" of the compounds disclosed herein include those derived from suitable bases such as alkali metals (e.g., sodium), alkaline earth metals (e.g., magnesium), ammonium, and NX 4 + (wherein X is C 1-4 Alkyl). Pharmaceutically acceptable salts of nitrogen atoms or amino groups include, for example, salts of: organic carboxylic acids, such as acetic acid, trifluoroacetic acid, adipic acid, and anti-bleedingAcid, aspartic acid, butyric acid, camphoric acid, cinnamic acid, citric acid, digluconic acid, glutamic acid, glycolic acid, glycerophosphoric acid, formic acid, caproic acid, benzoic acid, lactic acid, fumaric acid, tartaric acid, maleic acid, hydroxymaleic acid, malonic acid, malic acid, mandelic acid, isethionic acid, lactobionic acid, nicotinic acid, oxalic acid, pamoic acid, pectic acid, phenylacetic acid, 3-phenylpropionic acid, pivalic acid, propionic acid, pyruvic acid, salicylic acid, stearic acid, sulfanilic acid, tartaric acid, undecanoic acid, and succinic acid; organic sulfonic acids such as methanesulfonic acid, ethanesulfonic acid, camphorsulfonic acid, mesitylene sulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, naphthalenesulfonic acid, and 2-naphthalenesulfonic acid; and inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and sulfamic acid. Pharmaceutically acceptable salts of compounds of the hydroxyl group include anions of the compound with a suitable cation such as Na + And NX 4 + (wherein X is independently selected from H or C 1-4 Alkyl groups).
For therapeutic use, salts of the active ingredients of the compounds disclosed herein will generally be pharmaceutically acceptable, i.e., they are salts derived from physiologically acceptable acids or bases. However, pharmaceutically unacceptable salts of acids or bases may also be used, for example, to prepare or purify a compound of formula I or another compound of the embodiments disclosed herein. All salts (whether derived from physiologically acceptable acids or bases or not) are within the scope of the embodiments disclosed herein.
The metal salts are typically prepared by reacting a metal hydroxide with a compound according to embodiments disclosed herein. Examples of metal salts prepared in this way are those containing Li + 、Na + And K + Is a salt of (a). The less soluble metal salt may be precipitated from the more soluble salt solution by adding a suitable metal compound.
In addition, the salts may be formed from certain organic and inorganic acids (e.g., HCl, HBr, H 2 SO 4 、H 3 PO 4 Or organic sulfonic acid) with a basic center (typically an amine). Finally, it is to be understood that the compositions herein comprise an unionized form toAnd zwitterionic forms of the compounds disclosed herein.
"pharmaceutical composition" refers to a formulation of a compound of an embodiment disclosed herein and a medium commonly accepted in the art for delivery of a biologically active compound to a mammal (e.g., a human). Such vehicles include all pharmaceutically acceptable excipients.
An "effective amount" or "therapeutically effective amount" refers to an amount of a compound according to embodiments disclosed herein that is sufficient to effect treatment of a disease state, condition, or disorder disclosed herein when administered to a patient in need thereof. Such an amount will be sufficient to elicit a biological or medical response in the tissue system or in the patient sought by the researcher or clinician. The amount of a compound according to embodiments disclosed herein that constitutes a therapeutically effective amount will vary depending on factors such as: the compound and its biological activity, the composition used for administration, the time of administration, the route of administration, the rate of excretion of the compound, the duration of the treatment, the type of the disease state or disorder being treated and its severity, the drug combined with or used in combination with the compounds of the embodiments disclosed herein, and the age, weight, general health, sex, and diet of the patient. Such therapeutically effective amounts can be determined by one of ordinary skill in the art, given their own knowledge, prior art, and disclosure.
The term "treatment" as used herein is intended to mean administration of a compound or composition according to embodiments of the invention disclosed herein to reduce or eliminate one or more symptoms of HIV infection and/or to reduce viral load in a patient. In certain embodiments, the term "treating" also encompasses the administration of a compound or composition according to embodiments of the invention disclosed herein after exposure of an individual to a virus but before the occurrence of a disease symptom and/or before detection of a virus in the blood, to prevent the occurrence of a disease symptom and/or to prevent the virus from reaching a detectable level in the blood, and by administration to the mother prior to delivery and to the child within the first few days of life, to prevent perinatal transmission of HIV from the mother to the infant. The term "treating" also encompasses the administration of a compound or composition according to embodiments of the invention disclosed herein (also referred to as pre-exposure prevention or PrEP) prior to exposure of an individual to a virus to prevent the rooting of HIV infection and/or to prevent the virus from establishing a permanent infection and/or to prevent the occurrence of symptoms of a disease and/or to prevent the virus from reaching a detectable level in the blood when the individual is exposed to the virus. The term "treating" also encompasses the administration of a compound or composition according to embodiments of the invention disclosed herein both before and after exposure of an individual to a virus.
As used herein, the term "preventing" refers to administering a compound, composition or pharmaceutical salt according to the present disclosure before or after exposure of a human to a virus, but before the onset of symptoms of the disease and/or before the virus is detected in the blood. The term also refers to preventing the appearance of disease symptoms and/or preventing the virus from reaching a detectable level in the blood. The term includes pre-exposure prevention (PrEP), post-exposure prevention (PEP) and event-driven or "on-demand" prevention. The term also refers to preventing perinatal transmission of HIV from mother to infant by administration to the mother prior to delivery and to the child within the first few days of life. The term also refers to preventing transmission of HIV through blood infusion.
The term "antiviral agent" as used herein is intended to mean an agent (compound or biological agent) that is effective in inhibiting the formation and/or replication of a virus in a human, including but not limited to an agent that interferes with the host or viral machinery necessary for the formation and/or replication of a virus in a human.
The term "HIV replication inhibitor" as used herein is intended to mean an agent capable of reducing or eliminating the ability of HIV replication in a host cell (whether in vitro, ex vivo, or in vivo).
The compounds of the embodiments disclosed herein or their pharmaceutically acceptable salts may contain one or more asymmetric centers and thus may give rise to enantiomers, diastereomers and other stereoisomeric forms which may be defined as (R) -or (S) -or (D) -or (L) -for amino acids, depending on the absolute stereochemistry. The present disclosure is intended to include all such possible isomers, as well as their racemic, non-racemic, and optically pure forms. Optically active (+) and (-), (R) -and (S) -or (D) -and (L) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using methods such as chromatography and fractional crystallization. Techniques for preparing/separating the individual enantiomers include chiral synthesis from suitable optically pure precursors or resolution of the racemate (or of a salt or derivative) using, for example, chiral High Pressure Liquid Chromatography (HPLC). When a compound described herein contains an olefinic double bond or other geometric asymmetric center, and unless specified otherwise, it is intended that the compound include both E and Z geometric isomers. Also, all tautomeric forms are intended to be included.
"stereoisomers" refers to compounds that consist of the same atoms bonded by the same bonds but have different three-dimensional structures that are not interchangeable. The present disclosure contemplates various stereoisomers and mixtures thereof, and includes "enantiomers," which refers to two stereoisomers whose molecules are non-superimposable mirror images of each other. In any of the embodiments disclosed herein, the compounds disclosed herein can be in the form of stereoisomers thereof.
"partially unsaturated" refers to a cyclic group that contains at least one double bond but is not aromatic.
Substituents and multivalent groups can be attached to the remainder of the molecule at any position and in any orientation to produce stable compounds. For example, the number of the cells to be processed,wherein-X-Y-is defined as-CR 13A R 13B -CR 9 =CR 10 -a compound of formula I comprising formula->Compounds of formula (I)Is a compound of (a). Similarly, wherein-X-Y-is-CR 11A R 11B -CR 12A R 12B The compounds of formula I include the formula->Compounds of formula (I)Is a compound of (a).
II compounds
Disclosed herein are compounds of formula I:
or a pharmaceutically acceptable salt thereof, wherein
Ar is C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S; wherein said C 6 -C 10 Aryl or six to ten membered heteroaryl optionally being 1 to 4 independently selected from halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl and C 1 -C 6 Substituent substitution of the group consisting of alkoxy;
R 1 h, C of a shape of H, C 1 -C 3 Alkyl or phenyl;
R 2 is H or C 1 -C 3 An alkyl group;
R 3 is H or C 1 -C 3 An alkyl group;
R 4 and R is 5 Each independently is H, halogen, cyano, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S; wherein said C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or six-to ten-membered heteroaryl optionally being independently selected from halogen, C 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; or alternatively
R 4 And R is 5 Taken together form a 3-6 membered carbocyclic ring or a 4-6 membered heterocyclic ring containing one heteroatom selected from N, O and S;
R 6 is H, halogen or C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group;
R 7 is H, halogen or C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group;
R 8A and R is 8B Each independently H, C 1 -C 3 Alkyl or benzyl; and is also provided with
-X-Y-is- (CR) 13A R 13B ) p -CR 9 =CR 10 -or- (CR) 13A R 13B ) q -CR 11A R 11B -CR 12A R 12B -; wherein the method comprises the steps of
R 9 Is H, halogen or C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or C 1 -C 6 An alkoxy group;
R 10 is H, halogen or C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or C 1 -C 6 An alkoxy group; or alternatively
R 9 And R is 10 Together with the carbon to which they are attached, form a phenyl group or a 5-6 membered heteroaryl ring containing 1, 2 or 3 heteroatoms independently selected from N, O and S; wherein said phenyl or said 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 Substituent substitution of the group consisting of alkoxy; and is also provided with
R 11A 、R 11B 、R 12A 、R 12B 、R 13A And R is 13B Each independently is H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 An alkoxy group; or C 1 -C 6 A haloalkyl group; or alternatively
R 11A 、R 12A 、R 13A And R is 13B Each independently is H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 An alkoxy group; or C 1 -C 6 A haloalkyl group; and R is 11B And R is 12B Together with the carbon to which they are attached, form a 3-6 membered carbocyclic ring. Wherein the 3-6 membered carbocycle is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 Substituent substitution of the group consisting of alkoxy;
p is 0 or 1; and is also provided with
q is 0 or 1;
wherein when-X-Y is- (CR) 13A R 13B ) q -CR 11A R 11B -CR 12A R 12B -when, then:
(i)R 4 is halogen, cyano, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S; wherein said C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or six-to ten-membered heteroaryl optionally being independently selected from halogen, C 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; and R is 5 Is H, halogen, cyano, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl, or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S, wherein the C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or six-to ten-membered heteroaryl optionally being independently selected from halogen, C 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; or alternatively
(ii)R 4 And R is 5 Are linked together to form a 3-6 membered carbocyclic ring or a 4-6 membered heterocyclic ring having one heteroatom; or alternatively
(iii)R 8A Is C 1 -C 3 Alkyl or benzyl; or alternatively
(iv)R 6 Is halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group.
In some embodiments, the compounds of formula I provided herein have formula Ia:
in some embodiments, the compounds of formula I provided herein have formula Ib:
in some embodiments of the compounds of formula I, formula Ia and formula Ib, ar is C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S; wherein said C 6 -C 10 Aryl or six to ten membered heteroaryl groups are optionally substituted with 1-4 halogens. In some embodiments, ar is C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S; wherein said C 6 -C 10 Aryl or six to ten membered heteroaryl is optionally substituted with 1-3 substituents independently selected from Cl and F.
In some embodiments of the compounds of formula I, formula Ia and formula Ib, ar is C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one heteroatom selected from N, O and S; wherein said C 6 -C 10 Aryl or six to ten membered heteroaryl optionally being 1 to 4 independently selected from halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl and C 1 -C 6 The substituents of the group consisting of alkoxy groups. In some embodiments, ar is C 6 -C 10 Aryl or containing one hetero atom selected from N, O and SSix to ten membered heteroaryl; wherein said C 6 -C 10 Aryl or six to ten membered heteroaryl optionally being 1 to 4 independently selected from halogen, C 1 -C 6 Alkyl and C 1 -C 6 The substituents of the group consisting of alkoxy groups. In some embodiments, ar is C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one heteroatom selected from N, O and S; wherein said C 6 -C 10 Aryl or six to ten membered heteroaryl groups are optionally substituted with 1-4 halogens. In some embodiments, ar is C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one heteroatom selected from N, O and S; wherein said C 6 -C 10 Aryl or six to ten membered heteroaryl is optionally substituted with 1-3 substituents independently selected from Cl and F.
In some embodiments of the compounds of formula I, formula Ia and formula Ib, ar is optionally substituted with 1 to 4 groups independently selected from halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl and C 1 -C 6 Phenyl substituted by substituent groups of the group consisting of alkoxy. In some embodiments Ar is optionally substituted with 1 to 4 groups independently selected from halogen, C 1 -C 6 Alkyl and C 1 -C 6 Phenyl substituted by substituent groups of the group consisting of alkoxy. In some embodiments, ar is phenyl optionally substituted with 1 to 4 halogens. In some embodiments, ar is phenyl optionally substituted with 1 to 3 substituents independently selected from Cl and F.
In some embodiments of the compounds of formula I, formula Ia and formula Ib, ar is optionally substituted with one, two, three or four groups independently selected from halogen and C 1 -C 6 Phenyl substituted by substituent of alkoxy. In some embodiments, ar is independently selected from halo and C 1 -C 4 Phenyl substituted by substituent of alkoxy. In some embodiments, ar is phenyl substituted with one, two, three, or four substituents independently selected from Cl and F.
In some embodiments of the compounds of formula I, formula Ia, or formula Ib, ar is:
wherein Z is N or CR A
n is 0, 1, 2, 3 or 4; and is also provided with
Each R A Independently is halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl and C1-C 6 An alkoxy group.
In some embodiments of the compounds of formula I, formula Ia, and formula Ib, Z is CH or N. In some embodiments, Z is CH. In some embodiments, Z is N.
In some embodiments of the compounds of formula I, formula Ia and formula Ib, ar is n is 1, 2, 3 or 4; and each R A Independently is halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl and C 1 -C 6 An alkoxy group. In some embodiments of the compounds of formula I, formula Ia and formula Ib, ar is +.>n is 1, 2, 3 or 4; and each R A Independently is halogen, C 1 -C 6 Alkyl and C 1 -C 6 An alkoxy group. In some embodiments of the compounds of formula I, formula Ia and formula Ib, ar isn is 1, 2, 3 or 4; and each R A Independently halogen and C 1 -C 6 An alkoxy group. In some embodiments, ar is +.>n is 1, 2, 3 or 4; and each R A Independently halo and C 1 -C 4 An alkoxy group. In some embodimentsAr is->n is 1, 2 or 3; and each R A Independently fluorine or chlorine.
In some embodiments of the compounds of formula I, formula Ia and formula Ib, ar isn is 1, 2, 3 or 4; and each R A Independently is halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl and C 1 -C 6 An alkoxy group. In some embodiments of the compounds of formula I, formula Ia and formula Ib, ar is +.>n is 1, 2, 3 or 4; and each R A Independently is halogen, C 1 -C 6 Alkyl and C 1 -C 6 An alkoxy group. In some embodiments of the compounds of formula I, formula Ia and formula Ib, ar isn is 1, 2, 3 or 4; and each R A Independently halogen and C 1 -C 6 An alkoxy group. In some embodiments, ar is +. >n is 1, 2, 3 or 4; and each R A Independently halo and C 1 -C 4 An alkoxy group. In some embodiments, ar is +.>n is 1, 2 or 3; and each R A Independently fluorine or chlorine. />
In some embodiments of the compounds of formula I, formula Ia, or formula Ib, ar is
In some embodiments, a compound of formula I, formula Ia, or formula Ib disclosed herein has formula II:
wherein n is 0, 1, 2, 3 or 4; and each R A Independently is halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl and C 1 -C 6 An alkoxy group.
In some embodiments, the compounds of formula I, formula Ia, formula Ib, and formula II disclosed herein have formula IIa:
wherein n is 0, 1, 2, 3 or 4; and each R A Independently is halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl and C 1 -C 6 An alkoxy group.
In some embodiments, the compounds of formula I, formula Ib, and formula II disclosed herein have formula IIb:
wherein n is 0, 1, 2, 3 or 4; and each R A Independently is halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl and C 1 -C 6 An alkoxy group.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, -X-Y-is-CR 13A CR 13B -CR 9 =CR 10 -, each R is 9 、R 10 、R 13A And R is 13B Each independently of the otherIs H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or C 1 -C 6 An alkoxy group; or R is 13A And R is 13B Each independently is H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or C 1 -C 6 An alkoxy group; and R is 9 And R is 10 Together with the carbon to which they are attached, form a phenyl group or a 5-6 membered heteroaryl ring containing 1, 2 or 3 heteroatoms independently selected from N, O and S; wherein said phenyl or said 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, -X-Y-is-CR 9 =CR 10 -, each R is 9 And R is 10 Independently H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or C 1 -C 6 An alkoxy group; or R is 9 And R is 10 Together with the carbon to which they are attached, form a phenyl group or a 5-6 membered heteroaryl ring containing 1, 2 or 3 heteroatoms independently selected from N, O and S; wherein said phenyl or said 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, -X-Y-is-CH 2 -CR 9 =CR 10 -, wherein R is 9 And R is 10 Together with the carbon to which they are attached, form a phenyl group or a 5-6 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms independently selected from N, O and S. Wherein said phenyl or said 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups. In one placeIn some embodiments, -X-Y-is-CH 2 -CR 9 =CR 10 -, wherein R is 9 And R is 10 Together with the carbon to which they are attached, form a 5-6 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms independently selected from N, O and S; wherein the 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, -X-Y-is-CR 9 =CR 10 -, wherein R is 9 And R is 10 Together with the carbon to which they are attached, form a phenyl group or a 5-6 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms independently selected from N, O and S. Wherein said phenyl or said 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups. In some embodiments, -X-Y-is-CR 9 =CR 10 -, wherein R is 9 And R is 10 Together with the carbon to which they are attached, form a 5-6 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms independently selected from N, O and S; wherein the 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, -X-Y-is-CH 2 -CR 9 =CR 10 -, wherein R is 9 And R is 10 Together with the carbon to which they are attached, form a 5-6 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms independently selected from N, O and S; wherein the 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, -X-Y-is-CR 9 =CR 10 -, wherein R is 9 And R is 10 Together with the carbon to which they are attached, form a 5-6 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms independently selected from N, O and S; wherein the 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, -X-Y-is-CH 2 -CR 9 =CR 10 -,R 9 And R is 10 Together with the carbon to which they are attached, form a 5-6 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms independently selected from N and O; wherein the 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, -X-Y-is-CR 9 =CR 10 -,R 9 And R is 10 Together with the carbon to which they are attached, form a 5-6 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms independently selected from N and O; wherein the 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, R 11A 、R 11B 、R 12A 、R 12B 、R 13A And R is 13B Each independently is H, halogen, C 1 -C 6 Alkyl or C 1 -C 6 An alkoxy group. In some embodiments, R 11A 、R 11B 、R 12A 、R 12B 、R 13A And R is 13B Each independently is H, halogen or C 1 -C 6 An alkoxy group. In some embodiments, R 11A 、R 11B 、R 12A 、R 12B 、R 13A And R is 13B Each independently is H, halogen or methoxy. In some embodiments, R 11A 、R 11B 、R 12A 、R 12B 、R 13A And R is 13B Each independently is H, fluoro or methoxy.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, and formula IIb, there is formula III:
wherein the method comprises the steps of
m is 0, 1, 2 or 3; and is also provided with
Each R B Independently is halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or C 1 -C 3 An alkoxy group.
In some embodiments of the compounds of formula I, formula Ia, formula II, formula IIa, and formula III, having formula IIIa:
wherein the method comprises the steps of
m is 0, 1, 2 or 3; and is also provided with
Each R B Independently is halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or C 1 -C 3 An alkoxy group.
In some embodiments of the compounds of formula I, formula Ib, formula II, formula IIb, and formula III, having formula IIIb:
wherein the method comprises the steps of
m is 0, 1, 2 or 3; and is also provided with
Each R B Independently is halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or C 1 -C 3 An alkoxy group.
In some embodiments of compounds of formula III, formula IIIa and formula IIIb, m is 0, 1 or 2. In some embodiments, m is 0 or 1. In some embodiments, m is 1 or 2. In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3.
In some embodiments of the compounds of formula III, formula IIIa and formula IIIb, each R B Independently is halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 An alkoxy group. In some embodiments, each R B Independently is halogen, C 1 -C 3 Alkyl or C 1 -C 3 A haloalkyl group. In some embodiments, each R B Independently halogen or C 1 -C 3 An alkyl group. In some embodiments, each R B Independently halogen.
In some embodiments of the compounds of formula III, formula IIIa and formula IIIb, m is 0, 1 or 2, and each R B Independently is halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 An alkoxy group. In some embodiments, m is 0, 1 or 2, and each R B Independently is halogen, C 1 -C 3 Alkyl or C 1 -C 3 A haloalkyl group. In some embodiments, m is 0, 1 or 2, and each R B Independently halogen or C 1 -C 3 An alkyl group. In some embodiments, m is 0, 1 or 2, and each R B Independently halogen.
In some embodiments of compounds of formula III, formula IIIa and formula IIIb, m is 0 or 1, and each R B Independently is halogen, C 1 -C 3 Alkyl, C 1 -C 3 Halogenated compoundsAlkyl and C 1 -C 3 An alkoxy group. In some embodiments, m is 0 or 1, and each R B Independently is halogen, C 1 -C 3 Alkyl or C 1 -C 3 A haloalkyl group. In some embodiments, m is 0 or 1, and each R B Independently halogen or C 1 -C 3 An alkyl group. In some embodiments, m is 0 or 1, and each R B Independently halogen.
In some embodiments of compounds of formula III, formula IIIa and formula IIIb, m is 1 or 2, and each R B Independently is halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 An alkoxy group. In some embodiments, m is 1 or 2, and each R B Independently is halogen, C 1 -C 3 Alkyl or C 1 -C 3 A haloalkyl group. In some embodiments, m is 1 or 2, and each R B Independently halogen or C 1 -C 3 An alkyl group. In some embodiments, m is 1 or 2, and each R B Independently halogen.
In some embodiments of compounds of formula III, formula IIIa and formula IIIb, m is 1, and R B Is halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 An alkoxy group. In some embodiments, m is, and each R B Independently C 1 -C 3 Alkyl or C 1 -C 3 A haloalkyl group. In some embodiments, m is 1, and R B Is halogen or C 1 -C 3 An alkyl group. In some embodiments, m is 1, and R B Is halogen.
In some embodiments of compounds of formula III, formula IIIa and formula IIIb, m is 2, and each R B Independently is halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 An alkoxy group. In some embodiments, m is 2, and each R B Independently is halogen, C 1 -C 3 Alkyl or C 1 -C 3 A haloalkyl group. In some embodiments, m is 2, and each R B Independently halogen or C 1 -C 3 An alkyl group. In some embodiments, m is 2, and each R B Independently halogen.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, and formula IIb, there is formula IV:
Wherein z is 0 or 1.
In some embodiments of the compounds of formula I, formula Ia, formula II, formula IIa, and formula IV, having formula IVa:
wherein z is 0 or 1.
In some embodiments of the compounds of formula I, formula Ib, formula II, formula IIb, and formula IV, having formula IVb:
wherein z is 0 or 1.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, -X-Y-is-CR 9 =CR 10 -, each R is 9 And R is 10 Independently H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or C 1 -C 6 An alkoxy group. In some embodiments, -X-Y-is-CR 9 =CR 10 -, each R is 9 And R is 10 Independently H, halogen, C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group. In some embodiments, -X-Y-is-CR 9 =CR 10 -, each R is 9 And R is 10 Independently and separatelyIs H, halogen or C 1 -C 6 An alkyl group. In some embodiments, -X-Y-is-CR 9 =CR 10 -, each R is 9 And R is 10 Independently H or halogen. In some embodiments, -X-Y-is-CR 9 =CR 10 -, each R is 9 And R is 10 Independently H or F.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, -X-Y-is-CR 11A R 11B -CR 12A R 12B -, each R is 11A 、R 11B 、R 12A And R is 12B Independently H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group; or each R 11A And R is 12A Independently H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group; and R is 11B And R is 12B Together with the carbon to which they are attached, form a 3-6 membered carbocyclic ring. Wherein the 3-6 membered carbocycle is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, -X-Y-is-CR 11A R 11B -CR 12A R 12B -, each R is 11A 、R 11B 、R 12A And R is 12B Independently H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group. In some embodiments, -X-Y-is-CR 11A R 11B -CR 12A R 12B -, each R is 11A 、R 11B 、R 12A And R is 12B Independently H, halogen, C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group. In some embodiments, -X-Y-is-CR 11A R 11B -CR 12A R 12B -, each R is 11A 、R 11B 、R 12A And R is 12B Independently H, halogen or C 1 -C 6 An alkyl group. In some embodiments, -X-Y-is-CR 11A R 11B -CR 12A R 12B -, each R is 11A 、R 11B 、R 12A And R is 12B Independently H or halogen. In some embodiments, -X-Y-is-CR 11A R 11B -CR 12A R 12B -, each R is 11A 、R 11B 、R 12A And R is 12B Independently H or F. In some embodiments, -X-Y-is-CR 11A R 11B -CR 12A R 12B -, each R is 11A 、R 11B 、R 12A And R is 12B All are H.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa and formula IIb, -X-Y-is-CR 11A R 11B -CR 12A R 12B -, each R is 11A And R is 12A Independently H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group; and R is 11B And R is 12B Together with the carbon to which they are attached, form a 3-6 membered carbocyclic ring. Wherein the 3-6 membered carbocycle is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, and formula IIb, have formula V:
wherein z' is 1 or 2.
In some embodiments of the compounds of formula I, formula Ia, formula II, formula IIa, and formula V, have formula Va:
wherein z' is 1 or 2.
In some embodiments of the compounds of formula I, formula Ib, formula II, formula IIb, and formula V, have formula Vb:
wherein z' is 1 or 2.
In some embodiments of the compounds of formula V, formula Va, and formula Vb described herein, z' is 1. In some embodiments, z' is 2.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va and formula Vb described herein, R 8A Is H and R 8B H, C of a shape of H, C 1 -C 3 Alkyl or benzyl. In some embodiments, R 8A Is H and R 8B Is H or C 1 -C 3 An alkyl group. In some embodiments, R 8A Is H and R 8B H or benzyl. In some embodiments, R 8A Is H and R 8B Is C 1 -C 3 Alkyl or benzyl. In some embodiments, R 8A Is H and R 8B Is C 1 -C 3 An alkyl group. In some embodiments, R 8A Is and R is 8B Is benzyl. In some embodiments, R 8A And R is 8B All are H.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va and formula Vb described herein, R 8A Is C 1 -C 3 Alkyl and R 8B H, C of a shape of H, C 1 -C 3 Alkyl or benzyl. In some embodiments, R 8A Is C 1 -C 3 Alkyl and R 8B Is H or C 1 -C 3 An alkyl group. In one placeIn some embodiments, R 8A Is C 1 -C 3 Alkyl and R 8B H or benzyl. In some embodiments, R 8A Is C 1 -C 3 Alkyl and R 8B Is C 1 -C 3 Alkyl or benzyl. In some embodiments, R 8A Is C 1 -C 3 Alkyl and R 8B H. In some embodiments, R 8A Is C 1 -C 3 Alkyl and R 8B Is benzyl. In some embodiments, R 8A And R is 8B Are each independently C 1 -C 3 An alkyl group.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va and formula Vb described herein, R 8A Is benzyl and R 8B H, C of a shape of H, C 1 -C 3 Alkyl or benzyl. In some embodiments, R 8A Is benzyl and R 8B Is H or C 1 -C 3 An alkyl group. In some embodiments, R 8A Is benzyl and R 8B H or benzyl. In some embodiments, R 8A Is benzyl and R 8B Is C 1 -C 3 Alkyl or benzyl. In some embodiments, R 8A Is benzyl and R 8B Is C 1 -C 3 An alkyl group. In some embodiments, R 8A Is benzyl and R 8B H. In some embodiments, R 8A And R is 8B Are all benzyl groups.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va and formula Vb described herein, R 1 H, C of a shape of H, C 1 -C 3 Alkyl or phenyl and R 2 H. In some embodiments, R 1 Is H or phenyl and R 2 H. In some embodiments, R 1 Is H or C 1 -C 3 Alkyl and R 2 H. In some embodiments, R 1 Is C 1 -C 3 Alkyl or phenyl and R 2 H. In some embodiments, R 1 And R is 2 Both are H. In some embodiments, R 1 Is C 1 -C 3 Alkyl and R 2 H. In some embodiments, R 1 Is phenyl and R 2 H.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va and formula Vb described herein, R 1 H, C of a shape of H, C 1 -C 3 Alkyl or phenyl and R 2 Is C 1 -C 3 An alkyl group. In some embodiments, R 1 Is H or phenyl and R 2 Is C 1 -C 3 An alkyl group. In some embodiments, R 1 Is H or C 1 -C 3 Alkyl, and R 2 Is C 1 -C 3 An alkyl group. In some embodiments, R 1 Is C 1 -C 3 Alkyl or phenyl, and R 2 Is C 1 -C 3 An alkyl group. In some embodiments, R 1 Is H and R 2 Is C 1 -C 3 An alkyl group. In some embodiments, each R 1 And R is 2 Independently C 1 -C 3 An alkyl group. In some embodiments, R 1 Is phenyl and R 2 Is C 1 -C 3 An alkyl group. In some embodiments, each R 1 And R is 2 Independently H or C 1 -C 3 An alkyl group.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, and formula IVb described herein, each R 4 And R is 5 Independently H or C 1 -C 6 Alkyl, wherein C 1 -C 6 Alkyl groups optionally being independently selected from halogen, C by one, two or three 1 -C 3 Alkoxy or C 1 -C 3 The group substitution of haloalkoxy groups. In some embodiments, R 4 Is H or C 1 -C 6 Alkyl, wherein the C 1 -C 6 Alkyl groups optionallyOne, two or three of which are independently selected from halogen, C 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; and R is 5 Is H or C 1 -C 6 An alkyl group. In some embodiments, R 4 Is H or C 1 -C 6 Alkyl, wherein the C 1 -C 6 Alkyl groups optionally being independently selected from halogen, C by one, two or three 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; and R is 5 H.
In some embodiments of the compounds of formula IV, formula IVa and formula IVb described herein, R 4 Is halogen or C 1 -C 6 Alkyl, wherein C 1 -C 6 Alkyl groups optionally being independently selected from halogen, C by one, two or three 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; and R is 5 Is H, halogen or C 1 -C 6 Alkyl, wherein the C 1 -C 6 Alkyl groups optionally being independently selected from halogen, C by one, two or three 1 -C 3 Alkoxy or C 1 -C 3 The group substitution of haloalkoxy groups. In some embodiments, R 4 Is halogen or C 1 -C 6 Alkyl, wherein the C 1 -C 6 Alkyl groups optionally being independently selected from halogen, C by one, two or three 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; and R is 5 Is H, halogen or C 1 -C 6 An alkyl group. In some embodiments, R 4 Is halogen or C 1 -C 6 Alkyl, wherein the C 1 -C 6 Alkyl groups optionally being independently selected from halogen, C by one, two or three 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; and R is 5 H.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa and formula IVb, each R 4 And R is 5 Independently H, me, OMe or CH 2 F. In some embodiments, R 4 Is H, me, OMe or CH 2 F and R 5 H.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va and formula Vb described herein, R 4 And R is 5 Are linked together to form a 4-6 membered heterocyclic ring containing one heteroatom selected from N, O and S.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va and formula Vb described herein, R 4 And R is 5 Are linked together to form a 3-6 membered carbocyclic ring.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va and formula Vb described herein, R 6 Is H, halogen or C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group. In some embodiments, R 6 Is H, halogen or C 1 -C 6 An alkyl group. In some embodiments, R 6 Is halogen or C 1 -C 6 An alkyl group. In some embodiments, R 6 Is C 1 -C 6 An alkyl group. In some embodiments, R 6 H.
In some embodiments of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa and formula IVb described herein, R 7 Is H, halogen or C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group. In some embodiments, R 7 Is H, halogen or C 1 -C 6 An alkyl group. In some embodiments, R 7 Is halogen or C 1 -C 6 An alkyl group. In some embodiments, R 7 Is C 1 -C 6 An alkyl group. In some embodiments, R 7 H.
In some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is selected from the group consisting of:
/>
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is selected from the group consisting of:
/>
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is selected from the group consisting of:
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is selected from the group consisting of:
/>
/>
/>
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is selected from the group consisting of:
/>
/>
In some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is selected from:
/>
/>
/>
/>
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is selected from the group consisting of:
/>
/>
/>
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is selected from the group consisting of:
/>
/>
/>
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is selected from the group consisting of:
/>
/>
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is selected from the group consisting of:
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is:
In some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is:
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is:
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is:
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is:
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is:
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is:
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is:
In some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is:
/>
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is:
in some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb is:
III compositions and kits
The compounds provided herein are generally administered in the form of pharmaceutical compositions. Accordingly, also provided herein are pharmaceutical compositions comprising one or more compounds provided herein, or a pharmaceutically acceptable salt, isomer, or mixture thereof, and one or more pharmaceutically acceptable vehicles selected from carriers, adjuvants, and excipients. The compounds provided herein may be the only active ingredient or one of the active ingredients of the pharmaceutical composition. Suitable pharmaceutically acceptable vehicles may include, for example, inert solid diluents and fillers, diluents, including sterile aqueous solutions and various organic solvents, permeation enhancers, solubilizers, and adjuvants. Such compositions are prepared in a manner well known in the pharmaceutical arts. See, e.g., remington's Pharmaceutical Sciences, mace Publishing co., philiadelphia, pa., 17 th edition, (1985); and Modern Pharmaceutics, marcel Dekker, inc. 3 rd edition (edited by g.s. Banker and c.t. Rhodes).
In one aspect, provided herein are pharmaceutical compositions comprising a compound provided herein (e.g., a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb) or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient or carrier. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient or carrier.
In some embodiments, the pharmaceutical compositions provided herein further comprise one or more (e.g., one, two, three, four, one or two, one to three, or one to four) additional therapeutic agents or pharmaceutically acceptable salts thereof. In some embodiments, the pharmaceutical composition further comprises a therapeutically effective amount of one or more (e.g., one, two, three, four, one or two, one to three, or one to four) additional therapeutic agents or pharmaceutically acceptable salts thereof.
The pharmaceutical composition may be administered in single or multiple doses. The pharmaceutical compositions may be administered by a variety of methods including, for example, rectal, buccal, intranasal, and transdermal routes. In some embodiments, the pharmaceutical composition may be administered by intra-arterial injection, intravenous, intraperitoneal, parenteral, intramuscular, subcutaneous, oral, topical, or as an inhalant.
One mode of administration is parenteral, for example by injection. The pharmaceutical compositions described herein may be incorporated therein for administration by injection in forms such as aqueous or oil suspensions or emulsions, with sesame oil, corn oil, cottonseed oil or peanut oil, as well as elixirs, mannitol, dextrose or sterile aqueous solutions and similar pharmaceutical vehicles.
Oral administration may be another route for administering the compounds provided herein. Administration may be via, for example, a capsule or an enteric coated tablet. In preparing pharmaceutical compositions comprising at least one compound provided herein, or a pharmaceutically acceptable salt, isomer, or mixture thereof, an active ingredient (such as a compound provided herein) is typically diluted and/or encapsulated with an excipient within such a carrier, which may be in the form of a capsule, pouch, paper, or other container. When the excipient serves as a diluent, it may be in the form of a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the active ingredient. Thus, pharmaceutical compositions may be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, sterile injectable solutions and sterile packaged powders.
Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose or any combination thereof. The pharmaceutical composition may additionally include lubricants such as talc, magnesium stearate and mineral oil; a wetting agent; emulsifying and suspending agents; preservatives such as methyl benzoate and propyl hydroxybenzoate; a sweetener; and a flavoring agent; or any combination thereof.
Pharmaceutical compositions comprising at least one compound described herein, or a pharmaceutically acceptable salt, isomer, or mixture thereof, may be formulated so as to provide rapid, sustained, or delayed release of the active ingredient (such as the compounds provided herein) upon administration to a subject by employing procedures known in the art. Controlled release drug delivery systems for oral administration include osmotic pump systems and dissolution systems that include polymer-coated reservoirs or drug-polymer matrix formulations. Examples of controlled release systems are given in U.S. Pat. nos. 3,845,770, 4,326,525, 4,902,514 and 5,616,345. Another formulation for use in the methods of the present disclosure employs a transdermal delivery device ("patch"). Such transdermal patches may be used to provide continuous or discontinuous infusion of controlled amounts of the compounds provided herein. The construction and use of transdermal patches for delivering pharmaceutical agents is well known in the art. See, for example, U.S. Pat. nos. 5,023,252, 4,992,445 and 5,001,139. Such patches may be configured for continuous, pulsatile, or on-demand delivery of the medicament.
To prepare a solid composition (such as a tablet), the primary active ingredient may be admixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound described herein, or a pharmaceutically acceptable salt, isomer or mixture thereof. When these preformulated compositions are referred to as homogeneous, the active ingredient may be dispersed evenly throughout the composition so that the composition may be readily subdivided into equivalent unit dosage forms such as tablets, pills and capsules.
Tablets or pills of the compounds described herein can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action or protection from the acidic conditions of the stomach. For example, a tablet or pill may comprise an inner dosage component and an outer dosage component, the latter being in the form of an envelope over the former. The two components may be separated by an enteric layer that serves to resist disintegration in the stomach and allows the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials may be used for such enteric layers or coatings, including a variety of polymeric acids and mixtures of polymeric acids with materials such as shellac, cetyl alcohol and cellulose acetate.
Pharmaceutical compositions for inhalation or insufflation may comprise solutions and suspensions in pharmaceutically acceptable aqueous or organic solvents or mixtures thereof, as well as powders. The liquid or solid composition may contain suitable pharmaceutically acceptable excipients as described above. In some embodiments, the composition is administered by the oral or nasal respiratory route to achieve a local or systemic effect. In other embodiments, the composition in a pharmaceutically acceptable solvent may be nebulized by use of an inert gas. The aerosolized solution may be inhaled directly from the aerosolization device, or the aerosolization device may be attached to a mask tent or intermittent positive pressure ventilator. The solution, suspension or powder composition may be administered from a device that delivers the formulation in a suitable manner, preferably orally or nasally.
In one aspect, provided herein are kits comprising a compound provided herein (e.g., a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, and suitable packaging. In some embodiments, the kit further comprises instructions for use. In some embodiments, the kits include a label and/or instructions for use of a compound provided herein (e.g., a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb) or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, and a compound in treating an indication (including a disease or disorder) described herein.
In some embodiments, the kit further comprises one or more (e.g., one, two, three, four, one or two, one to three, or one to four) additional therapeutic agents or pharmaceutically acceptable salts thereof.
In one aspect, provided herein are articles of manufacture comprising a compound described herein, or a pharmaceutically acceptable salt, isomer, or mixture thereof, in a suitable container. In some embodiments, the container may be a vial, a jar, an ampoule, a prefilled syringe, or an intravenous bag.
IV method
In one embodiment, a method of treating HIV infection in a human having or at risk of HIV (e.g., HIV-1 and/or HIV-2) infection is provided, the method comprising administering to the human a therapeutically effective amount of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb.
In some embodiments, the method further comprises administering to the human a therapeutically effective amount of one, two, three, or four additional therapeutic agents. In certain embodiments, the one or more additional therapeutic agents are anti-HIV agents. In particular embodiments, the one or more additional therapeutic agents are HIV protease inhibitors, non-nucleoside or non-nucleotide inhibitors of HIV reverse transcriptase, nucleoside or nucleotide inhibitors of HIV reverse transcriptase, HIV capsid inhibitors, gp41 inhibitors, CXCR4 inhibitors, gpl20 inhibitors, CCR5 inhibitors, latency reversing agents, capsid polymerization inhibitors, HIV bNAb (broadly neutralizing HIV antibodies), TLR7 agonists, pharmacokinetic enhancers, other drugs for treating HIV, or combinations thereof. In one embodiment, the one or more additional therapeutic agents is abacavir, tenofovir alafenamide, tenofovir disoproxil, lenacipam, or a pharmaceutically acceptable salt thereof. In one embodiment, the one or more additional therapeutic agents is abacavir, tenofovir alafenamide, tenofovir disoproxil, lenacipam, GS-5894, isala Qu Wei, or a pharmaceutically acceptable salt thereof. In some embodiments, the one or more additional therapeutic agents is lenacipam, eosla Qu Wei. In some embodiments, the additional therapeutic agent is lenacipam. In some embodiments, the additional therapeutic agent is eosla Qu Wei.
In another embodiment, there is provided the use of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, for the treatment of HIV infection in a human suffering from or at risk of HIV (e.g., HIV-1 and/or HIV-2) infection.
In another embodiment, a pharmaceutical composition of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, or a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof is provided for use in medical therapy.
In another embodiment, a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, is provided for use in the treatment of HIV infection.
In another embodiment, a pharmaceutical composition of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, or a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, is provided for use in treating an HIV infection in a human suffering from or at risk of suffering from an HIV infection.
In another embodiment, there is provided a pharmaceutical composition of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, or a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, for use in a method of treating HIV infection in a human having or at risk of HIV infection, wherein the method further comprises administering one, two, three, or four additional therapeutic agents to the human.
In another embodiment, there is provided a pharmaceutical composition of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, or a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, for use in a method of treating HIV infection in a human suffering from or at risk of HIV infection, wherein the method further comprises administering to the human one, two, three, or four additional therapeutic agents selected from the group consisting of: HIV protease inhibitors, non-nucleoside or non-nucleotide inhibitors of HIV reverse transcriptase, nucleoside or nucleotide inhibitors of HIV reverse transcriptase, HIV capsid inhibitors, gp41 inhibitors, CXCR4 inhibitors, gpl20 inhibitors, CCR5 inhibitors, latency reversers, capsid polymerization inhibitors, HIV bNAb, TLR7 agonists, pharmacokinetic enhancers, other drugs for treating HIV, or combinations thereof. In one embodiment, one, two, three or four additional therapeutic agents are selected from the group consisting of HIV protease inhibitors, non-nucleoside inhibitors of HIV reverse transcriptase, nucleoside or nucleotide inhibitors of HIV reverse transcriptase, latency reversing agents, HIV capsid inhibitors, HIV bNAb, TLR7 agonists, and combinations thereof.
In another embodiment, there is provided a pharmaceutical composition for treating HIV infection of a human having or at risk of having an HIV infection, a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, or a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, wherein the method further comprises administering a therapeutically effective amount of tenofovir dipivoxil and emtricitabine to the human.
In another embodiment, there is provided a pharmaceutical composition of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, or a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, for use in a method of treating HIV infection in a human having or at risk of HIV infection, wherein the method further comprises administering to the human a therapeutically effective amount of tenofovir dipivoxil and emtricitabine.
In another embodiment, there is provided a pharmaceutical composition of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, or a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, for use in a method of treating HIV infection in a human having or at risk of HIV infection, wherein the method further comprises administering to the human a therapeutically effective amount of tenofovir dipivoxil.
In another embodiment, a pharmaceutical composition for treating HIV infection of a human having or at risk of having an HIV infection is provided, wherein the method further comprises administering to the human a therapeutically effective amount of tenofovir alafenamide.
In another embodiment, a method of using a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb in therapy is provided. In particular, a method of treating HIV viral proliferation, treating AIDS, or delaying the onset of AIDS or ARC symptoms in a mammal (e.g., a human) is provided, comprising administering to the mammal a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
In another embodiment, a composition comprising a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient is provided for use in a method of treating HIV viral proliferation, treating AIDS, or delaying the onset of AIDS or ARC symptoms in a mammal (e.g., a human).
In one embodiment, a pharmaceutical composition of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, or a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof is provided for use in preventing HIV infection.
For example, in one embodiment, a pharmaceutical composition of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, or a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, is provided for use in pre-exposure prophylaxis (prip), i.e., in preventing viral infection rooting upon exposure of an individual to HIV virus and/or preventing the onset of a permanent infection of the virus and/or preventing the appearance of symptoms of the disease and/or preventing the virus from reaching a detectable level in blood, prior to exposure to an HIV virus.
In another embodiment, the use of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating HIV infection in a human suffering from or at risk of HIV infection is disclosed.
In another embodiment, the use of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof, as a research tool is disclosed.
In another embodiment, an article of manufacture is disclosed comprising a composition effective to treat HIV infection; and packaging material comprising a label indicating that the composition can be used to treat infection by HIV. Exemplary compositions comprise a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt thereof.
In another embodiment, a method of inhibiting HIV replication is disclosed. The method comprises exposing the virus to an effective amount of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va or formula Vb or a salt thereof under conditions that inhibit HIV replication.
In another embodiment, the use of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va or formula Vb to inhibit HIV integrase activity is disclosed.
In another embodiment, disclosed is the use of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a salt thereof, to inhibit HIV replication.
V. application of
The compounds of the present disclosure (also referred to herein as active ingredients) may be administered by any route suitable for the condition to be treated. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), transdermal, vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), and the like. It will be appreciated that the preferred route may vary depending on, for example, the recipient's conditions. An advantage of certain compounds disclosed herein is that they are orally bioavailable and can be administered orally.
The compounds of the present disclosure may be administered to an individual for a desired period or duration of time, such as at least about 1 month, at least about 2 months, at least about 3 months, at least about 6 months, or at least about 12 months or longer, according to an effective dosing regimen. In some embodiments, the compound is administered on a daily or intermittent schedule during the life of the individual.
The specific dosage level of a compound of the present disclosure for any particular subject will depend on a variety of factors including the activity of the particular compound employed, the age, weight, general health, sex, diet, time of administration, route of administration and rate of excretion, drug combination and the severity of the particular disease of the subject undergoing therapy. For example, the dosage may be expressed as milligrams of the compound described herein (mg/kg) per kilogram of subject body weight. Dosages between about 0.1mg/kg and 150mg/kg may be appropriate. In some embodiments, a dosage of between about 0.1mg/kg and 100mg/kg may be appropriate. In other embodiments, a dosage of between 0.5mg/kg and 60mg/kg may be appropriate. Normalization according to the weight of a subject is particularly useful in adjusting the dose between subjects of widely varying sizes, such as when using drugs in children and adults, or when converting an effective dose in a non-human subject such as a dog to a dose suitable for a human subject.
Daily doses can also be described as the total amount of a compound described herein per dose or administered daily. The daily dose of a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb, or a pharmaceutically acceptable salt or pharmaceutically acceptable tautomer thereof, may be between about 1mg and 4,000mg, between about 2,000 and 4,000 mg/day, between about 1 and 2,000 mg/day, between about 1 and 1,000 mg/day, between about 10 and 500 mg/day, between about 20 and 500 mg/day, between about 50 and 300 mg/day, between about 75 and 200 mg/day, or between about 15 and 150 mg/day.
The dosage or frequency of administration of the compounds of the present disclosure can be adjusted during the course of treatment at the discretion of the administering physician.
The compounds of the present disclosure can be administered to an individual (e.g., a human) in a therapeutically effective amount. In some embodiments, the compound is administered once daily.
The compounds provided herein can be administered by any useful route and means, such as by oral or parenteral (e.g., intravenous) administration. A therapeutically effective amount of a compound may include from about 0.00001mg/kg body weight/day to about 10mg/kg body weight/day, such as from about 0.0001mg/kg body weight/day to about 10mg/kg body weight/day, or such as from about 0.001mg/kg body weight/day to about 1mg/kg body weight/day, or such as from about 0.01mg/kg body weight/day to about 1mg/kg body weight/day, or such as from about 0.05mg/kg body weight/day to about 0.5mg/kg body weight/day. In some embodiments, a therapeutically effective amount of a compound provided herein comprises from about 0.3 mg/day to about 30 mg/day, or from about 30 mg/day to about 300 mg/day, or from about 0.3 μg/day to about 30 mg/day, or from about 30 μg/day to about 300 μg/day.
The compounds of the present disclosure may be combined with one or more additional therapeutic agents at any dose of the compounds of the present disclosure (e.g., 1mg to 1000mg of the compound). A therapeutically effective amount may comprise from about 0.1 mg/dose to about 1000 mg/dose, such as from about 50 mg/dose to about 500 mg/dose, or such as from about 100 mg/dose to about 400 mg/dose, or such as from about 150 mg/dose to about 350 mg/dose, or such as from about 200 mg/dose to about 300 mg/dose, or such as from about 0.01 mg/dose to about 1000 mg/dose, or such as from about 0.01 mg/dose to about 100 mg/dose, or such as from about 0.1 mg/dose to about 100 mg/dose, or such as from about 1 mg/dose to about 10 mg/dose, or such as from about 1 mg/dose to about 1000 mg/dose. Other therapeutically effective amounts of the compounds of formula I, formula Ia, formula Ib, formula II, formula IIa, formula IIb, formula III, formula IIIa, formula IIIb, formula IV, formula IVa, formula IVb, formula V, formula Va, or formula Vb are about 1 mg/dose, or about 2 mg/dose, 3 mg/dose, 4 mg/dose, 5 mg/dose, 6 mg/dose, 7 mg/dose, 8 mg/dose, 9 mg/dose, 10 mg/dose, 15 mg/dose, 20 mg/dose, 25 mg/dose, 30 mg/dose, 35 mg/dose, 40 mg/dose, 45 mg/dose, 50 mg/dose, 55 mg/dose, 60 mg/dose, 65 mg/dose, 70 mg/dose, 75 mg/dose, 80 mg/dose, 85 mg/dose, 90 mg/dose, 95 mg/dose, or about 100 mg/dose. Other therapeutically effective amounts of the compounds of the present disclosure are about 100 mg/dose, 125 mg/dose, 150 mg/dose, 175 mg/dose, 200 mg/dose, 225 mg/dose, 250 mg/dose, 275 mg/dose, 300 mg/dose, 325 mg/dose, 350 mg/dose, 375 mg/dose, 400 mg/dose, 425 mg/dose, 450 mg/dose, 475 mg/dose, 500 mg/dose, 525 mg/dose, 550 mg/dose, 575 mg/dose, 600 mg/dose, 625 mg/dose, 650 mg/dose, 675 mg/dose, 700 mg/dose, 725 mg/dose, 750 mg/dose, 775 mg/dose, 800 mg/dose, 825 mg/dose, 850 mg/dose, 875 mg/dose, 900 mg/dose, 925 mg/dose, 950 mg/dose, 975 mg/dose, or about 1000 mg/dose per dose.
In some embodiments, the methods described herein comprise administering to the subject an initial daily dose of about 1mg to 500mg of a compound herein, and gradually increasing the dose until clinical efficacy is achieved. Increments of about 5mg, 10mg, 25mg, 50mg or 100mg may be used to increase the dose. The dosage may be increased daily, every other day, twice weekly, biweekly, tricyclically or monthly.
When administered orally, the total daily dose of a human subject may be between about 1mg and 1,000mg, between about 10 mg/day and 500 mg/day, between about 50 mg/day and 300 mg/day, between about 75 mg/day and 200 mg/day, or between about 100 mg/day and 150 mg/day. In some embodiments, the total daily dose of a human subject may be about 100 mg/day, 200 mg/day, 300 mg/day, 400 mg/day, 500 mg/day, 600 mg/day, 700 mg/day, 800 mg/day, 900 mg/day, or 1000 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 200 mg/day, 300 mg/day, 400 mg/day, 500 mg/day, 600 mg/day, 700 mg/day, or 800 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 300 mg/day, 400 mg/day, 500 mg/day, or 600 mg/day administered in a single dose.
In some embodiments, the total daily dose of a human subject may be about 100 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 150 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 200 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 250 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 300 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 350 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 400 mg/day administered in a single dose. In some embodiments, the total daily dose of the human subject may be about 450 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 500 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 550 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 600 mg/day administered in a single dose. In some embodiments, the total daily dose of the human subject may be about 650 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 700 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 750 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 800 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 850 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 900 mg/day administered in a single dose. In some embodiments, the total daily dose of the human subject may be about 950 mg/day administered in a single dose. In some embodiments, the total daily dose of a human subject may be about 1000 mg/day administered in a single dose.
A single dose may be administered hourly, daily, weekly or monthly. For example, a single dose may be administered every 1 hour, 2 hours, 3 hours, 4 hours, 6 hours, 8 hours, 12 hours, 16 hours, or every 24 hours. A single dose may also be administered once every 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, or once every 7 days. A single dose may also be administered once every 1 week, 2 weeks, 3 weeks, or once every 4 weeks. In certain embodiments, a single dose may be administered once a week. A single dose may also be administered once a month. In some embodiments, the compounds disclosed herein are administered once daily in the methods disclosed herein. In some embodiments, the compounds disclosed herein are administered twice daily in the methods disclosed herein.
In some embodiments, the compounds disclosed herein are administered once every 10 days. In some embodiments, the compounds disclosed herein are administered once every 15 days. In some embodiments, the compounds disclosed herein are administered once every 20 days. In some embodiments, the compounds disclosed herein are administered every 10-15 days. In some embodiments, the compounds disclosed herein are administered every 15-20 days. In some embodiments, the compounds disclosed herein are administered every 10-20 days. In some embodiments, the compounds disclosed herein are administered once per month. In some embodiments, the compounds disclosed herein are administered once every 2 months. In some embodiments, the compounds disclosed herein are administered once every 3 months. In some embodiments, the compounds disclosed herein are administered once every 4 months. In some embodiments, the compounds disclosed herein are administered once every 5 months. In some embodiments, the compounds disclosed herein are administered once every 6 months. In some embodiments, the compounds disclosed herein are administered once every 8 months. In some embodiments, the compounds disclosed herein are administered once every 10 months. In some embodiments, the compounds disclosed herein are administered once a year.
The frequency of the dosage of the compounds of the present disclosure will be determined by the needs of the individual patient and may be, for example, once a day or two or more times a day. Administration of the compound continues for a period of time required to treat HBV infection, HIV infection, cancer, hyperproliferative disease, or any other indication described herein. For example, the compound may be administered to a person infected with HBV for a period of 20 to 180 days, or for example, for a period of 20 to 90 days, or for example, for a period of 30 to 60 days.
Administration may be intermittent, with the patient receiving a daily dose of a compound of the present disclosure over a period of days or more, followed by a period of days or more, without the patient receiving a daily dose of the compound. For example, the patient may receive a dose of the compound every other day or three times a week. Again by way of example, a patient may receive a dose of compound daily for a period of 1 day to 14 days, then the patient does not receive a dose of compound for a period of 7 days to 21 days, then the patient receives a dose of compound again for a subsequent period of time (e.g., 1 day to 14 days). Alternate periods of administration of the compound followed by no administration of the compound may be repeated depending on the clinical needs of the patient being treated.
The compounds of the present disclosure or pharmaceutical compositions thereof may be administered once, twice, three times, or four times a day using any suitable pattern described above. Moreover, administration or treatment with the compound may continue for a number of days; for example, for one treatment cycle, typically treatment will continue for at least 7 days, 14 days, or 28 days. The treatment cycle is well known in cancer chemotherapy, typically alternating with a rest period of about 1 to 28 days, typically about 7 days or about 14 days between cycles. In other embodiments, the treatment cycle may also be continuous.
VI combination therapy
In certain embodiments, a method is provided for treating or preventing HIV infection in a human having or at risk of having an HIV infection, comprising administering to the human a therapeutically effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, in combination with a therapeutically effective amount of one or more (e.g., one, two, three, one, or two or one to three) additional therapeutic agents. In one embodiment, a method is provided for treating HIV infection in a human having or at risk of having HIV infection, comprising administering to the human a therapeutically effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, in combination with a therapeutically effective amount of one or more (e.g., one, two, three, one, or two or one to three) additional therapeutic agents.
In one embodiment, a pharmaceutical composition is provided comprising a compound disclosed herein, or a pharmaceutically acceptable salt thereof, in combination with one or more (e.g., one, two, three, one, or two or one to three) additional therapeutic agents, and a pharmaceutically acceptable carrier, diluent, or excipient.
In certain embodiments, the present disclosure provides a method for treating HIV infection comprising administering to a patient in need thereof a therapeutically effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, in combination with a therapeutically effective amount of one or more additional therapeutic agents suitable for treating HIV infection.
In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with one, two, three, four, or more additional therapeutic agents. In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with two additional therapeutic agents. In other embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with three additional therapeutic agents. In further embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with four additional therapeutic agents. The additional therapeutic agents may be different therapeutic agents selected from the same class of therapeutic agents, and/or they may be selected from different classes of therapeutic agents.
Administration of HIV combination therapies
In certain embodiments, the compounds disclosed herein are administered with one or more additional therapeutic agents. Co-administration of a compound disclosed herein with one or more additional therapeutic agents generally refers to simultaneous or sequential administration of a compound disclosed herein and one or more additional therapeutic agents such that a therapeutically effective amount of both the compound disclosed herein and the one or more additional therapeutic agents are present in the patient. When administered sequentially, the combination may be administered in two or more administrations.
Co-administration includes administration of a unit dose of a compound disclosed herein before or after administration of a unit dose of one or more additional therapeutic agents. For example, the compounds disclosed herein may be administered within seconds, minutes, or hours of administration of one or more additional therapeutic agents. In some embodiments, a unit dose of a compound disclosed herein is administered first, followed by administration of the unit dose of one or more additional therapeutic agents within seconds or minutes. Alternatively, a unit dose of one or more additional therapeutic agents is administered first, followed by administration of a unit dose of a compound disclosed herein within seconds or minutes. In other embodiments, a unit dose of a compound disclosed herein is administered first, followed by administration of the unit dose of one or more additional therapeutic agents after several hours (e.g., 1 hour to 12 hours). In further embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed by administration of a unit dose of a compound disclosed herein after several hours (e.g., 1 hour to 12 hours).
In certain embodiments, the compounds disclosed herein are combined with one or more additional therapeutic agents in a single dosage form for simultaneous administration to a patient, e.g., as a solid dosage form for oral administration.
In certain embodiments, the compounds of formula I are formulated as tablets, which may optionally contain one or more other compounds useful in the treatment of HIV. In certain embodiments, the tablet may contain another active ingredient for the treatment of HIV, such as an HIV protease inhibitor, a non-nucleoside or non-nucleotide inhibitor of HIV reverse transcriptase, a nucleoside or nucleotide inhibitor of HIV reverse transcriptase, an HIV integrase inhibitor, an HIV non-catalytic site (or allosteric) integrase inhibitor, a pharmacokinetic enhancer, and combinations thereof.
In certain embodiments, such tablets are suitable for once-a-day administration.
Combination therapy for HIV
In the above embodiments, the additional therapeutic agent may be an anti-HIV agent. HIV protease inhibitors, non-nucleoside or non-nucleotide inhibitors of HIV reverse transcriptase, nucleoside or nucleotide inhibitors of HIV reverse transcriptase, HIV integrase inhibitors, HIV non-catalytic site (or allosteric) integrase inhibitors, HIV entry inhibitors, HIV maturation inhibitors, immunomodulators, immunotherapeutic agents, antibody-drug conjugates, gene modifiers, gene editors (such as CRISPR/Cas9, zinc finger nucleases, homing nucleases, synthetic nucleases, TALENs), cell therapies (such as chimeric antigen receptor T-cell CAR-T and engineered T-cell receptor TCR-T, autologous T cell therapies), latency reversers, HIV capsid targeting compounds, immune-based therapies, phosphatidylinositol 3-kinase (PI 3K) inhibitors, HIV antibodies, bispecific antibodies and "antibody-like" therapeutic proteins HIV p17 matrix protein inhibitors, IL-13 antagonists, peptidyl prolyl cis-trans isomerase A modulators, protein disulfide isomerase inhibitors, complement C5a receptor antagonists, DNA methyltransferase inhibitors, HIV Vif gene modulators, vif dimerization antagonists, HIV-1 viral infectious agent inhibitors, TAT protein inhibitors, HIV-1Nef modulators, hck tyrosine kinase modulators, mixed lineage kinase-3 (MLK-3) inhibitors, HIV-1 splice inhibitors, rev protein inhibitors, integrin antagonists, nucleoprotein inhibitors, splice factor modulators, COMM domain-containing protein 1 modulators, CD4 antagonists, HIV ribonuclease H inhibitors, defensin modulators, CDK-9 inhibitors, CCR5 chemokine antagonists, CCR5 gene modulators, dendritic ICAM-3 capture non-integrin 1 inhibitors, HIV GAG protein inhibitors, HIV POL protein inhibitors, hyaluronidase inhibitors, nef antagonists, nef inhibitors, protease activated receptor-1 antagonists, tnfα ligand inhibitors, PDE4 inhibitors, complement factor H modulators, ubiquitin ligase inhibitors, deoxycytidine kinase inhibitors, cyclin dependent kinase inhibitors, proprotein convertase PC9 stimulators, ATP dependent RNA helicase DDX3X inhibitors, reverse transcriptase priming complex inhibitors, G6PD and NADH oxidase inhibitors, pharmacokinetic enhancers, HIV gene therapy, HIV vaccines, and combinations thereof.
In some embodiments, the additional therapeutic agent is selected from the group consisting of: combination drugs for HIV, other drugs for the treatment of HIV, HIV protease inhibitors, HIV reverse transcriptase inhibitors, HIV integrase inhibitors, HIV non-catalytic site (or allosteric) integrase inhibitors, HIV entry (fusion) inhibitors, HIV maturation inhibitors, latency reversers, capsid inhibitors, immune based therapies, PI3K inhibitors, HIV antibodies and bispecific antibodies and "antibody-like" therapeutic proteins, and combinations thereof.
HIV combination medicine
Examples of combination drugs include(efavirenz, tenofovir disoproxil fumarate and emtricitabine); />(bictevir, emtricitabine and tenofovir alafenamide); />(Rilpivirine, tenofovir disoproxil fumarate and emtricitabine); />(eptifibatide, cobicistat, tenofovir disoproxil fumarate, and emtricitabine); />(tenofovir disoproxil fumarate)And emtricitabine; tdf+ftc); />(tenofovir alafenamide and emtricitabine);(tenofovir alafenamide, emtricitabine, and rilpivirine); />(tenofovir alafenamide, emtricitabine, cobicistat, and etiracer); / >(darunavir, tenofovir alafenamide hemifumarate, emtricitabine, and cobalastat); SYMFITM (efavirenz, lamivudine and tenofovir disoproxil fumarate); CIMDUTM (lamivudine and tenofovir disoproxil fumarate); tenofovir and lamivudine; tenofovir alafenamide and emtricitabine; tenofovir alafenamide hemifumarate and emtricitabine; tenofovir alafenamide hemifumarate, emtricitabine and rilpivirine; tenofovir alafenamide hemifumarate, emtricitabine, cobicistat and eptifibatide; />(zidovudine and lamivudine; azt+3tc); />(/>Abacavir sulfate and lamivudine; abc+3tc); />(/>Lopinavir and ritonavirA (2) a (v);(dolutegravir, abacavir, and lamivudine); />(abacavir sulfate, zidovudine, and lamivudine; abc+azt+3tc); atazanavir and cobalastat; atazanavir sulfate and cobalastat; atazanavir sulfate and ritonavir; darunavir and cobalastat; dolutegravir and rilpivirine; dolutegravir and rilpivirine hydrochloride; dolutegravir, abacavir sulfate, and lamivudine; lamivudine, nevirapine and zidovudine; raltegravir and lamivudine; doravirine, lamivudine, and tenofovir disoproxil fumarate; doravirine, lamivudine and tenofovir disoproxil; dapivirine+levonorgestrel, dulutetravir+lamivudine, dulutetravir+emtricitabine+tenofovir alafenamide, ai Fawei lin+emtricitabine+tenofovir disoproxil, lamivudine+abacavir+zidovudine, lamivudine+abacavir, lamivudine+tenofovir disoproxil fumarate, lamivudine+zidovudine+nevirapine, lopinavir+ritonavir, lopinavir+ritonavir+abacavir+lamivudine, lopinavir+zidovudine+lamivudine, tenofovir dipivoxil fumarate+emtricitabine+rilpivirine hydrochloride, lopinavir, ritonavir, zidovudine and lamivudine.
Other HIV drugs
Examples of other drugs for the treatment of HIV include acetylmorpholine, alisporivir, atovam, banLec, CC-11050, deferiprone, gamimune, griffonian, mi Tefa, naltrexone, prolastin, REP 9, RPI-MN, vorapaxar, VSSP, H1viral, SB-728-T, 1, 5-dicaffeoylquinic acid, rHIV7-shl-TAR-CCR5RZ, mazF gene therapy, MK-8527, blockAide, PSC-RANTES, ABX-464, AG-1105, APH-0812, BIT-225, CYT-107, HGTV-43, HPH-116, HS-10234, IMO-3100, IND-02, MK-1376, MK-2048, MK-4250, MK-8507, MK-8591, NOV-205, PA-1050040 (PA-040), PGN-007, SCY-635, SB-9200, SCB-452, APH-0812, BIT-225, CYT-107, MK-1376, MK-2048, MK-90111, 9018 and 90mu.113.
HIV protease inhibitors
Examples of HIV protease inhibitors include amprenavir, atazanavir, bei Kana (brecanavir), darunavir, fosamprenavir calcium, indinavir sulfate, lopinavir, nelfinavir mesylate, ritonavir, saquinavir mesylate, telanavir, DG-17, TMB-657 (PPL-100), T-169, BL-008, MK-8122, TMB-607 and TMC-310911.
HIV reverse transcriptase inhibitors
Examples of non-nucleoside or non-nucleotide inhibitors of HIV reverse transcriptase include dapivirine, delavirdine mesylate, doravirine, efavirenz, itravirenz, lentinan (lentinan), MK-8583, nevirapine, rilpivirine, TMC-278LA, ACC-007, AIC-292, KM-023, PC-1005 and Es Sha Weilin (VM-1500).
Examples of nucleoside or nucleotide inhibitors of HIV reverse transcriptase include adefovir, adefovir dipivoxil, albefrane, albefradine, emtricitabine, tenofovir alafenamide tenofovir alafenamide fumarate, tenofovir alafenamide hemifumarate, tenofovir disoproxil fumarate tenofovir disoproxil fumarate, tenofovir disoproxil hemifumarate,And VIDEX->(didanosine, ddl), abacavir sulfate, alovudine, aliscitabine, cinnafdine, didanosine, elvucitabine, fetenavir, fosfoglite, CMX-157, dapivirine, doravirine, itravirrine, OCR-5753, tenofovir disoproxil orotate,Fuzivudine, exenatide (islastrovir), lamivudine, azidothaphos, stavudine, zalcitabine, zidovudine, luo Wafu, i.e., etafenamide (GS-9131), GS-9148, MK-8504, MK-8591, MK-858, VM-2500, and KP-1461./ >
HIV integrase inhibitors
Examples of HIV integrase inhibitors include ivermectin Lei Wei, curcumin, derivatives of curcumin, chicoric acid, derivatives of 3, 5-dicaffeoylquinic acid, aurintricarboxylic acid, derivatives of aurintricarboxylic acid, phenethyl caffeate, derivatives of phenethyl caffeate, tyrosine kinase inhibitors, derivatives of tyrosine kinase inhibitors, quercetin, derivatives of quercetin, raltegravir, dolutevir, JTK-351, bicaprevir, AVX-15567, BMS-986197, cabspecific (long-acting injection), dikeoquinoline-4-1 derivatives, integrase-LEDGF inhibitors, leggins, M-522, M-532, NSC-310217, NSC-371056, NSC-48240, NSC-642710, NSC-699171, NSC-699172, NSC-699173, NSC-699174, stilbenedisulfonic acid, T-169, and VM-3500.
Examples of HIV non-catalytic sites or allosteric integrase inhibitors (NCINI) include CX-05045, CX-05168, and CX-14442.
HIV entry inhibitors
Examples of HIV entry (fusion) inhibitors include cenicriviroc, CCR5 inhibitors, gp41 inhibitors, CD4 attachment inhibitors, DS-003 (BMS-599793), gp120 inhibitors and CXCR4 inhibitors.
Examples of CCR5 inhibitors include alavir, valvirrol, maravirrol, cenicriviroc, leronelimab (PRO-140), adatavir (RAP-101), nifevirol (TD-0232), anti-GP 120/CD4 or CCR5 bispecific antibodies, B-07, MB-66, polypeptide C25P, TD-0680 and vMIP (haiminipu).
Examples of gp41 inhibitors include Ebolovide Tay, enfuvirtide, BMS-986197, enfuvirtide bio-improvers, enfuvirtide bio-imitation, HIV-1 fusion inhibitor (P26-Bapc), ITV-1, ITV-2, ITV-3, ITV-4, PIE-12 trimer and Wesevirtide.
Examples of inhibitors of CD4 attachment include eba Li Zhushan antibodies and CADA analogs.
Examples of gp120 inhibitors include Radha-108 (acceptor alcohol) 3B3-PE38, banLec, bentonite-based nanomedicine, foster Sha Weian butanetriol, ilp-0831 and BMS-663068.
Examples of CXCR4 inhibitors include pleshafu, ALT-1188, N15 peptide and vMIP (Haimipu).
HIV maturation inhibitors
Examples of HIV maturation inhibitors include BMS-955176, BMS-986197, GSK-3640254 and GSK-2838232.
Latency reversing agent
Examples of latency reversing agents include Histone Deacetylase (HDAC) inhibitors, proteasome inhibitors (such as velcade and ifer Sha Zuo meter citrate), protein Kinase C (PKC) activators, smyd2 inhibitors, BET-bromodomain 4 (BRD 4) inhibitors, ionomycin, PMA, SAHA (suberoylanilide hydroxamic acid or suberoylanilide, aniline and hydroxamic acid), IL-15 modulating antibodies, JQ1, disulfiram, amphotericin B and ubiquitin inhibitors (such as lagzolla analogs) and APH-0812 and GSK-343.
Examples of HDAC inhibitors include romidepsin, vorinostat, and panobinostat.
Examples of PKC activators include indololactam, prostratin, ingenol B and DAG-lactone.
Capsid inhibitor
Examples of capsid inhibitors include capsid polymerization inhibitors or capsid disruption compounds, HIV core capsid p7 (NCp 7) inhibitors (such as azodicarbonamide), HIV p24 capsid protein inhibitors, GS-6207, AVI-621, AVI-101, AVI-201, AVI-301, and AVI-CAN1-15 families.
Immunization-based therapies
Examples of immune-based therapies include toll-like receptor modulators such as tlr1, tlr2, tlr3, tlr4, tlr5, tlr6, tlr7, tlr8, tlr9, tlr10, tlr11, tlr12, and tlr13; a regulator of apoptosis protein 1 (Pd-1); a programmed death ligand 1 (Pd-L1) modulator; IL-15 modulators; dermaVir; interleukin-7; plaquenil (hydroxychloroquine); prometryne (aldinterleukin, IL-2); interferon alpha; interferon alpha-2 b; interferon alpha-n 3; pegylated interferon alpha; interferon gamma; hydroxyurea; mycophenolate Mofetil (MPA) and its ester derivative Mycophenolate Mofetil (MMF); ribavirin; polymeric Polyethylenimine (PEI); a gepon; IL-12; WF-10; VGV-1; MOR-22; BMS-936559; CYT-107, interleukin-15/Fc fusion protein, AM-0015, ALT-803, NIZ-985, NKTR-255, NKTR-262, NKTR-214, novolone, pegylated interferon alpha-2 a, pegylated interferon alpha-2 b, recombinant interleukin-15, xmab-24306, RPI-MN, STING modulator, RIG-I modulator, NOD2 modulator, SB-9200 and IR-103.
Examples of TLR agonists: dimension Sha Mode (GS-9620), GS-986, IR-103, diphenhydramine, tilsotolimod, runtatimod, DSP-0509, AL-034, G-100, cobitlimod, AST-008, mo Tuo molod, GSK-1795091, GSK-2245035, VTX-1463, GS-9688, LHC-165, BDB-001, RG-7854, telratolimod. RO-7020531.
Phosphatidylinositol 3-kinase (PI 3K) inhibitors
Examples of PI3K inhibitors include Ai Deli sibirinib, albo Li Xibu (alpelinib), bupirinib (buparliib), orotic acid CAI, colupib, du Weili sibirinib, gada Li Xibu (gedatoliib), lenatinib, panolinib Li Xibu (panulinib), pirifaxine, idalixib (piciliib), pilapinib Li Xibu (pilaliib), prasugrel quinidine mesylate, regatinib sodium Sonolisib, tassentib, AMG-319, AZD-8186, BAY-1082439, CLR-1401, CLR-457, CUDC-907, DS-7423, EN-3342, GSK-2126458, GSK-2269577, GSK-2636771, INCB-040093, LY-3023414, MLN-1117, PQR-309, RG-7666, RP-6530, RV-1729, SAR-245409, SAR-260301, SF-1126, TGR-1202, UCB-5857, VS-5584, XL-765 and ZTK-474.
Alpha-4/beta-7 antagonists
Examples of integrin alpha-4/beta-7 antagonists include PTG-100, TRK-170, A Li Lushan antibodies, E Qu Lizhu mAb, methylcaroglib (carotegrast methyl) and vedolizumab.
HIV antibodies, bispecific antibodies and "antibody-like" therapeutic proteins
Examples of HIV antibodies, bispecific antibodies, and "antibody-like" therapeutic proteins include Fab derivatives, bispecific antibodies, trispecific antibodies, multivalent antibodies, bnAB (broadly neutralizing HIV-1 antibodies), BMS-936559, TMB-360 and those antibodies targeting HIV GP120 or GP41, antibody recruiting molecules targeting HIV, anti-CD 63 monoclonal antibodies, CD3 bispecific antibodies, CD16 bispecific antibodies, anti-GB virus C antibodies, anti-GP 120/CD4, CCR5 bispecific antibodies, anti-Nef single domain antibodies, anti-Rev antibodies, camelid-derived anti-CD 18 antibodies, camelid-derived anti-ICAM-1 antibodies, DCVax-001, GP140 targeting antibodies, GP 41-based HIV therapeutic antibodies, human recombinant mAb (PGT-121), ebalizumab, immuglo, MB-66.
Examples of those antibodies that target HIV in this manner include baveltuximab, UB-421, C2F5, 2G12, C4E10, C2F5+C2G12+C4E10, 8ANC195, 3BNC117-LS, 3BNC60, D1D2, 10-1074-LS, GS-9722, DH411-2, BG18, PGT145, PGT121, PGT122, PGT-151, PGT-133, PGT-135, PGT-128, MDX010 (ipilimab), DH511-2, N6LS, N49P6, N49P7, N49P7.1, N49P9, N49P11 N60P1.1, N60P25.1, N60P2.1, N60P31.1, N60P22, NIH 45-46, PG9, PG16, 8ANC195, 2Dm2m, 4Dm2m, 6Dm2m, VRC-01-LS, PGDM1400, A32, 7B2, 10E8VLS, 3810109, 10E8v4, 10E8.4/iMab, VRC-01/PGDM-1400/10E8v4, IMC-HIV, iMabm36, 10E8v4/PGT121-VRC01, eCD4-Ig, IOMA, CAP256-VRC26.25, DRVIA7, SAR-441236, VRC-07-523, VRC-HIAB 080-00-AB, VRC-HIVMAB060-00-AB, P2G12 and VRC07. Examples of HIV bispecific antibodies include MGD014, TMB bispecific antibodies.
Examples of in vivo delivered bnAB such as AAV8-VRC07, mRNA encoding anti-HIV antibody VRC 01.
Pharmacokinetic enhancers
Examples of pharmacokinetic enhancers include cobalamin and ritonavir.
Additional therapeutic agents
Examples of additional therapeutic agents include compounds in WO 2004/096286 (Gilead Sciences), WO 2006/015261 (Gilead Sciences), WO 2006/110157 (Gilead Sciences), WO 2012/003497 (Gilead Sciences), WO 2012/003498 (Gilead Sciences), WO 2012/145728 (Gilead Sciences), WO 2013/006738 (Gilead Sciences), WO 2013/159064 (Gilead Sciences), WO 2014/100323 (Gilead Sciences), US 3/0165489 (University of Pennsylvania), US 2014/0221378 (Japan Tobacco), US 2014/0221380 (Japan Tobacco), WO 2009/062285 (Boehringer Ingelheim), WO 2010/130034 (Boehringer Ingelheim), WO 2013/792 (Gilead Sciences), US 20140221356 (Gilead Sciences), US 20100143301 (Gilead Sciences), and US 1093/0923.
HIV vaccine
Examples of HIV vaccines include peptide vaccines, recombinant subunit protein vaccines, live vector vaccines using viral vectors such as arenavirus, lymphocytic choriomeningitis virus (LCMV), pi Qinde virus, modified vaccinia ankara virus (MVA), adenovirus, adeno-associated virus (AAV), vesicular Stomatitis Virus (VSV) and chimpanzee adenovirus (ChAd), DNA vaccines, CD4 derived peptide vaccines, vaccine combinations, BG505 sosip.664gp140, rgp120 (AIDSVAX), ALVAC HIV (vCP 1521)/AIDSVAX B/E (GP 120) (144), monomeric GP120 HIV-subtype 1C vaccine, remune, ITV-1, content Vir, ad4-Env145NFL Ad5-ENVA-48, HB-500, DCVax-001 (CDX-2401), vacc-4x, vacc-C5, vacc-CRX, VVX-004, VAC-3S, multistage DNA recombinant adenovirus-5 (rAD 5), rAD5 Gag-pol Env A/B/C vaccine, pennvax-G, pennvax-GP/MVA-CMDR, HIV-Trimix-mRNA vaccine, HIV-LAMP-vax, ad35-GRIN, NAcGM3/VSSP ISA-51, polyICLC adjuvanted vaccine, tatImmune, GTU-mutiplHIV (FIT-06), GP140[ delta ] V2.TV1+MF-59, rVSVIN HIV-1Gag vaccine, seV-Gag vaccine, AT-20, DNK-4, ad35-Grin/ENV, TBC-M4, HIX, HIVAX-2, NYHIV-1, NYYHIV-PT-123, HIV-PT-123, VAC-PT-123, rAAV1-PG9DP, GOVX-B11, GOVX-B21, TVI-HIV-1, ad-4 (Ad 4-Env Clade C+Ad 4-mGAG), paxvax, EN41-UGR7C, EN-FPA 2, preVaxTat, AE-H, MYM-V101, combiHIVvac, ADVAX, MYM-V201, MVA-CMDR, DNa-Ad5gag/pol/nef/nev (HVTN 505), MVATG-17401, ETV-01, CDX-1401, rcAD26.MOS1.HIV-Env, ad26.Mod. HIV vaccine, ad26.Mod. HIV+MVA mosaic vaccine+gp 140, AGS-004, AVX-101, AVX-201, PEP-6409, SAV-001, thV-01, TUTI-16, VGX-3300, VGV-001 and virus-like particle (such as 37B), 37B-24. MOV-like particle, GTU-like vaccine, DNA-like fusion vaccine HIV gag/pol/nef/Env DNA vaccine, anti-TAT HIV vaccine, conjugated polypeptide vaccine, dendritic cell vaccine, gag-based DNA vaccine, GI-2010, GP41 HIV-1 vaccine, HIV vaccine (PIKA adjuvant), I i-key/MHC class II epitope hybrid peptide vaccine, ITV-2, ITV-3, ITV-4, LIPO-5, multistage Env vaccine, MVA vaccine, pennvax-GP, pp 71-deficient HCMV vector HIV gag vaccine, recombinant peptide vaccine (HIV infection), NCI, rgp160 HIV vaccine, RNActive HIV vaccine, SCB-703, TAT Oyi vaccine, TBC-M4, therapeutic HIV vaccine, UBI HIV GP120, vacc-4x+romidepsin, variant GP120 polypeptide vaccine, rAD5 gag-pol Env A/B/C vaccine, DNA.HTI, DNA.HTI and MVA.HTI, VRC-HIVDNA016-00-VP+VRC-HIVADV014-00-VP, INO-6145, JNJ-9220, gp145 C.6980; eOD-GT8 60mer based vaccines, PD-201401, env (A, B, C, A/E)/gag (C) DNA vaccines, gp120 (A, B, C, A/E) protein vaccines, PDPHV-201401, ad4-EnvCN54, envseq-1 Envs HIV-1 vaccine (GLA-SE adjuvant), HIV p24gag prime-boost plasmid DNA vaccines, arenavirus vector based immunotherapy (Vaxwave, theraT), MVA-BN HIV-1 vaccine regimens, MVA.tHIVconsv4, MVA.tHIVconsv3, UBI HIV gp120, mRNA based prophylactic vaccines, TBL-1203HI, VRC-HIVR096-00-VP, VAX-3S, HIV MAG DNA vaccines.
Combination therapy for HIV
In a specific embodiment, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with one, two, three, four, or more additional therapeutic agents selected from the group consisting of:(efavirenz, tenofovir disoproxil fumarate and emtricitabine); />(/>Rilpivirine, tenofovir disoproxil fumarate and emtricitabine); />(eptifibatide, cobicistat, tenofovir disoproxil fumarate, and emtricitabine); />(tenofovir disoproxil fumarate and emtricitabine; TDF+FTC);(tenofovir alafenamide and emtricitabine); />(tenofovir alafenamide, emtricitabine, and rilpivirine); />(tenofovir alafenamide, emtricitabine, cobicistat, and etiracer); adefovir; adefovir dipivoxil; cocoa butter; emtricitabine; tenofovir; tenofovir disoproxil; fumaric acidTenofovir disoproxil; tenofovir alafenamide; tenofovir alafenamide hemifumarate; />(dolutegravir, abacavir, and lamivudine); dolutegravir, abacavir sulfate, and lamivudine; raltegravir; raltegravir and lamivudine; maraviroc; enfuwei peptide; / >(/>Lopinavir and ritonavir);(zidovudine and lamivudine; azt+3tc); />(/>Abacavir sulfate and lamivudine; abc+3tc); />(abacavir sulfate, zidovudine, and lamivudine; abc+azt+3tc); rilpivirine; rilpivirine hydrochloride; atazanavir sulfate and cobalastat; atazanavir and cobalastat; darunavir and cobalastat; atazanavir; atazanavir sulfate; dolutegravir; entecavir; ritonavir; atazanavir sulfate and ritonavir; darunavir is used; lamivudine; pravastatin; fosamprenavir; fosamprenavir calcium efavirenz; itravirin; nelfinavir is used; nelfinavir mesylate; an interferon; didanosine; stavudine; indinavir; indinavir sulfate; tenofovir and lamivudine; zidovudine; nevirapine; saquinavir; saquinavir mesylate; aldesleukin; zalcitabine; telanavir; amprenavir; delafuSetting; delavirdine mesylate; radha-108 (acceptor alcohol); lamivudine and tenofovir disoproxil fumarate; efavirenz, lamivudine and tenofovir disoproxil fumarate; azido phosphine; lamivudine, nevirapine and zidovudine; abacavir; and abacavir sulfate.
Those skilled in the art will appreciate that the additional therapeutic agents listed above may be included in more than one of the categories listed above. The specific classes are not intended to limit the function of those compounds listed in those classes.
In a specific embodiment, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with an HIV reverse transcriptase nucleoside or nucleotide inhibitor and an HIV reverse transcriptase non-nucleoside inhibitor. In another specific embodiment, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with an HIV reverse transcriptase nucleoside or nucleotide inhibitor and an HIV protease inhibiting compound. In further embodiments, the compounds disclosed herein, or a pharmaceutically acceptable salt thereof, are combined with an HIV reverse transcriptase nucleoside or nucleotide inhibitor, an HIV reverse transcriptase non-nucleoside inhibitor, and a pharmacokinetic enhancer. In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with at least one HIV nucleoside inhibitor, integrase inhibitor, and pharmacokinetic enhancer of reverse transcriptase. In another embodiment, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with two HIV nucleoside or nucleotide inhibitors of reverse transcriptase.
In a specific embodiment, a compound disclosed herein or a pharmaceutically acceptable salt thereof is combined with abacavir sulfate, tenofovir disoproxil fumarate, tenofovir alafenamide hemifumarate, or tenofovir alafenamide hemifumarate.
In a specific embodiment, a compound disclosed herein or a pharmaceutically acceptable salt thereof is combined with tenofovir, tenofovir disoproxil fumarate, tenofovir alafenamide or tenofovir alafenamide hemifumarate.
In a specific embodiment, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with a first additional therapeutic agent selected from the group consisting of abacavir sulfate, tenofovir disoproxil fumarate, tenofovir alafenamide and tenofovir alafenamide hemifumarate and a second additional therapeutic agent selected from the group consisting of emtricitabine and lamivudine.
In a specific embodiment, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with a first additional therapeutic agent selected from the group consisting of tenofovir, tenofovir disoproxil fumarate, tenofovir alafenamide, and tenofovir alafenamide hemifumarate, and a second additional therapeutic agent, wherein the second additional therapeutic agent is emtricitabine.
A compound as disclosed herein (e.g., any compound of formula I) may be combined with one or more additional therapeutic agents at any dose of a compound of formula I (e.g., 1mg to 500mg of a compound).
In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with 5mg to 30mg tenofovir alafenamide fumarate, tenofovir alafenamide hemifumarate, or tenofovir alafenamide and 200mg emtricitabine. In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with 5mg to 10mg, 5mg to 15mg, 5mg to 20mg, 5mg to 25mg, 25mg to 30mg, 20mg to 30mg, 15mg to 30mg, or 10mg to 30mg tenofovir alafenamide fumarate, tenofovir alafenamide hemifumarate, or tenofovir alafenamide, and 200mg emtricitabine. In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with 10mg tenofovir alafenamide fumarate, tenofovir alafenamide hemifumarate, or tenofovir alafenamide and 200mg emtricitabine. In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with 25mg tenofovir alafenamide fumarate, tenofovir alafenamide hemifumarate, or tenofovir alafenamide and 200mg emtricitabine. Compounds as disclosed herein (e.g., compounds of formula I) can be combined with the agents provided herein in any dose of compounds (e.g., 1mg to 500mg of compounds), as if each combination was specifically and individually listed in doses.
In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with 200mg to 400mg tenofovir disoproxil fumarate, tenofovir disoproxil hemifumarate, or tenofovir disoproxil fumarate and 200mg emtricitabine. In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with 200mg to 250, 200mg to 300, 200mg to 350, 250mg to 400, 350mg to 400, 300mg to 400, or 250mg to 400mg tenofovir disoproxil fumarate, tenofovir disoproxil hemi-fumarate, or tenofovir disoproxil fumarate and 200mg emtricitabine. In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with 300mg of tenofovir disoproxil fumarate, or tenofovir disoproxil fumarate and 200mg of emtricitabine. Compounds as disclosed herein (e.g., compounds of formula I) can be combined with the agents provided herein in any dose of compounds (e.g., 1mg to 500mg of compounds), as if each combination was specifically and individually listed in doses.
In one embodiment, a kit is provided that includes a compound disclosed herein, or a pharmaceutically acceptable salt thereof, in combination with one or more (e.g., one, two, three, one, or two or one to three) additional therapeutic agents.
Contraceptive (contraceptive) combination therapy
Therapeutic agents (contraceptives) for controlling birth include cyproterone acetate, desogestrel, dienogest, drospirenone, estradiol valerate, ethinyl estradiol, norgestrel, etogestrel, levomethyltetrahydrofolate, levonorgestrel, lynetrogestone, medroxyprogesterone acetate, ethinyl estrol methyl ether, mifepristone, misoprostol, nomestrone acetate, norgestrel, norgestimate, omexifene, selenoacetate (segestersone acetate), ulipristal acetate, and any combination thereof.
Gene therapy and cell therapy
Gene therapy and cell therapy include genetic modification of a silenced gene; genetic methods for direct killing of infected cells; infusion of immune cells designed to replace the immune system of most patients themselves to enhance the immune response to the infected cells, or activate the immune system of patients themselves to kill the infected cells, or to find and kill the infected cells; genetic methods of modifying cellular activity to further alter the endogenous immune response to infection.
Examples of dendritic cell therapies include AGS-004.
Examples of CCR5 gene editing drugs include, for example, SB-728T.
Examples of inhibitors of the CCR5 gene include, for example, cal-1.
CD4 positive T cells expressing C34-CCR5/C34-CXCR 4.
AGT-103 transduced autologous T cell therapy.
AAV-eCD4-Ig gene therapy.
Gene edit
The genome editing system is selected from the group consisting of: CRISPR/Cas9 systems, zinc finger nuclease systems, TALEN systems, homing endonuclease systems, and meganuclease systems.
Examples of HIV-targeted CRISPR/Cas9 systems include EBT-101.
CAR-T cell therapy
The population of immune effector cells is engineered to express a Chimeric Antigen Receptor (CAR), wherein the CAR comprises an HIV antigen binding domain. HIV antigens include HIV envelope protein or portion thereof, gp120 or portion thereof, CD4 binding site on gp120, CD 4-induced binding site on gp120, N-glycans on gp120, V2 on gp120, membrane proximal region on gp 41. The immune effector cells are T cells or NK cells. In some embodiments, the T cell is a cd4+ T cell, a cd8+ T cell, or a combination thereof. The cells may be autologous or allogeneic.
Examples of HIV CAR-T include VC-CAR-T, anti-CD 4 CART cell therapy, autologous hematopoietic stem cells genetically engineered to express CD4 CAR and C46 peptide.
TCR-T cell therapy
TCR-T cells are engineered to target HIV-derived peptides present on the surface of virus-infected cells.
Examples VII. Examples
Exemplary chemical entities of the present disclosure are provided in the following specific examples. Those skilled in the art will recognize that in order to obtain the various compounds herein, the starting materials may be appropriately selected such that the final desired substituents will be reacted, optionally with or without protection, to yield the desired product. Alternatively, it may be necessary or desirable to replace the final desired substituent with a suitable group that can be subjected to the reaction scheme and optionally substituted with the desired substituent. Furthermore, one of skill in the art will recognize that the transformations shown in the schemes below may be performed in any order compatible with the functionality of the particular pendant group.
The examples provided herein describe the synthesis of the compounds disclosed herein and intermediates useful in the preparation of these compounds. It should be understood that the various steps described herein may be combined. It will also be appreciated that individual batches of the compounds may be combined and then continued in the next synthesis step.
In the following example description, specific embodiments are described. These embodiments are described in sufficient detail to enable those skilled in the art to practice certain embodiments of the disclosure. Other embodiments may be utilized, and logical and other changes may be made without departing from the scope of the present disclosure. Accordingly, the following description is not intended to limit the scope of the present disclosure.
Examples 1 to 4: preparation
(6R) -10-fluoro-1-hydroxy-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13- Methanobenzo [ g ] bridged]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (C1)
(6R) -9-fluoro-1-hydroxy-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13- Methanobenzo [ g ] bridged]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (C2)
(6S) -10-fluoro-1-hydroxy-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13- Methanobenzo [ g ] bridged]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (C3)
(6S) -9-fluoro-1-hydroxy-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13- Methanobenzo [ g ] bridged]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (C4)
5- (benzyloxy) -2, 3-dihydro-1H-pyrido [2,1-f ] [1,2,4] triazine-4, 6-dione (1) was prepared according to the procedure disclosed in WO 2019/160683 A1 for "intermediate A".
Step 1: preparation of 1- (benzyloxy) -10-fluoro-7, 12-dihydro-6, 13-methanobenzo [ g ]]Pyrido [1,2 ] b][1,2,5]Triazacyclononane-2, 14-dione (2-a) and 1- (benzyloxy) -9-fluoro-7, 12-dihydro-6, 13-methano Benzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-2, 14-dione (2-b)
5- (benzyloxy) -2, 3-dihydro-1H-pyrido [2,1-f ][1,2,4]Triazine-4, 6-dione (1) (200 mg,0.737 mmol) and 1, 2-bis (bromomethyl) -4-fluorobenzene (229 mg, 0.81mmol) were mixed with DMF (5 ml) and the mixture was cooled to 0 ℃. NaOt-Bu (159 mg,1.66 mmol) was added over 1 hour. The reaction mixture was allowed to stir overnight without reloading into a cold bath. The reaction mixture was then usedEtOAc diluted with NH 4 Cl/water treatment. The organic phase was separated and concentrated. The residue was purified by silica gel chromatography with 0-100% EtOAc in heptane and then 10% MeOH in EtOAc to give a mixture of the two positional isomers of the product (2-a and 2-b). MS (m/z): 392.1[ M+H ]]. The two positional isomers were not separated and the next step was continued.
Step 2: preparation of 1- (benzyloxy) -10-fluoro-3-iodo-7, 12-dihydro-6, 13-methanobenzo [ g ]]Pyrido-s [1,2-b][1,2,5]Triazacyclononane-2, 14-dione (3-a) and 1- (benzyloxy) -9-fluoro-3-iodo-7, 12-dihydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-2, 14-dione (3-b)
1- (benzyloxy) -10-fluoro-7, 12-dihydro-6, 13-methanobenzo [ g ] at room temperature]Pyrido [1,2-b][1,2,5]Triazacyclononane-2, 14-dione (2-a) and 1- (benzyloxy) -9-fluoro-7, 12-dihydro-6, 13-methanobenzo [ g ] ]Pyrido [1,2-b][1,2,5]A mixture of triazacyclononane-2, 14-dione (2-b) (50 mg,0.128 mmol) was admixed with MeOH (0.8 ml). m-CPBA (77%) (114 mg,0.51 mmol) and NIS (114 mg,0.51 mmol) were added sequentially. The reaction vial was sealed and heated from room temperature to 80 ℃ for 30 minutes. m-CPBA (77%) (114 mg,0.51 mmol) and NIS (114 mg,0.51 mmol) were then added sequentially. The reaction mixture was again heated at 80 ℃ for 30 minutes. The reaction mixture was then diluted with EtOAc and taken up in NaHCO 3 Water and Na 2 S 2 O 3 (10%) aqueous solution treatment. The organic phase was separated and concentrated. The residue was purified on a silica gel column with 0-100% EtOAc/heptane to give 60mg of the product as a mixture of the two positional isomers (3-a and 3-b). MS (m/z): 518.06[ M+H ]]. The two positional isomers were not separated and the next step was continued.
Step 3: preparation: (6R) -1- (benzyloxy) -10-fluoro-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7, 12, 14-tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (4-a)
(6R) -1- (benzyloxy) -9-fluoro-2, 14-dioxo-N- (2, 4, 6-trifluoro)Benzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (4-b)
(6S) -1- (benzyloxy) -10-fluoro-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro- - 6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (4-c)
(6S) -1- (benzyloxy) -9-fluoro-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (4-d)
To 1- (benzyloxy) -10-fluoro-3-iodo-7, 12-dihydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-2, 14-dione (3-a) and 1- (benzyloxy) -9-fluoro-3-iodo-7, 12-dihydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]To a solution of triazacyclononane-2, 14-dione (3-b) (32 mg,0.062 mmol) in DMSO (2 ml) was added 2,4, 6-trifluorobenzylamine (50 mg,0.309 mmol), DIPEA (40 mg,0.309 mmol) and Pd (PPh) 3 ) 4 (3.57 mg,0.00309 mmol). The reaction mixture was bubbled with CO (g) for 10 minutes. The reaction mixture was then heated at 80 ℃ under CO atmosphere for 17 hours. The reaction mixture was cooled to room temperature and diluted with ethyl acetate. The resulting mixture was treated with 0.05N HCl. The organic phase was separated and treated with saturated sodium bicarbonate solution and brine. The organic phase is then taken up in Na 2 SO 4 Drying and concentrating. The residue was purified by column on silica gel with 0% -100% etoac in heptane to give the desired product. SFC separation of this mixture of four isomers (ADH 50 IPA-NH) 3 ) The 4 isomers were obtained in ascending order of retention time: (6R) -1- (benzyloxy) -10-fluoro-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]10mg of triazacyclononane-3-carboxamide (4-a). MS (m/z): 579.08[ M+H ]]The method comprises the steps of carrying out a first treatment on the surface of the (6R) -1- (benzyloxy) -9-fluoro-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (4-b) 6mg。MS(m/z):579.08[M+H]The method comprises the steps of carrying out a first treatment on the surface of the (6S) -1- (benzyloxy) -10-fluoro-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (4-c) 7mg. MS (m/z): 579.03[ M+H ]]The method comprises the steps of carrying out a first treatment on the surface of the And (6S) -1- (benzyloxy) -9-fluoro-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (4-d) 4mg. MS (m/z): 579.02[ M+H ]]。
Step 4: preparation of (6R) -10-fluoro-1-hydroxy-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14- Tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (C1)
(6R) -1- (benzyloxy) -10-fluoro-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g ] at room temperature ]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (4-a) (10 mg,0.0173 mmol) was dissolved in toluene (0.5 ml). TFA (0.5 ml) was added in one portion. The reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was concentrated to dryness. The residue was dissolved in MeOH and taken up in 0.1% TFA in 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilization afforded the product as a TFA salt. MS (m/z): 489.26[ M+H ]] +1 H NMR (400 MHz, acetonitrile-d 3) delta 10.13 (s, 1H), 8.52 (s, 1H), 7.41-7.22 (m, 1H), 7.13 (d, j=9.3 hz, 1H), 7.04 (t, j=8.6 hz, 1H), 6.88 (t, j=8.6 hz, 2H), 5.56 (d, j=16.7 hz, 1H), 4.87-4.69 (m, 2H), 4.64 (d, j=5.7 hz, 2H), 4.55 (d, j=15.3 hz, 2H), 4.13 (d, j=13.2 hz, 1H).
Preparation of (6R) -9-fluoro-1-hydroxy-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (C2)
The synthesis of the title product was carried out in the same manner as in (C1) except that (6R) -9-fluoro-1-hydroxy-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (4-b) as starting material instead of (6R) -1- (benzyloxy) -10-fluoro-2, 14-dioxo -N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Except for triazacyclononane-3-carboxamide (4-a). The product was obtained as TFA salt. MS (m/z): 489.14[ M+H ]] +1 H NMR (400 MHz, acetonitrile-d 3) delta 10.14 (s, 1H), 8.54 (d, j=17.3 hz, 1H), 7.36 (dd, j=8.4, 5.7hz, 1H), 7.23-6.98 (m, 2H), 6.88 (t, j=8.5 hz, 2H), 5.51 (d, j=16.5 hz, 1H), 4.81 (p, j=14.7, 14.0hz, 2H), 4.64 (d, j=5.4 hz, 2H), 4.56 (dd, j=15.0, 10.4hz, 2H), 4.20 (d, j=13.6 hz, 1H).
Preparation of (6S) -10-fluoro-1-hydroxy-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro- 6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (C3)
The synthesis of the title product was carried out in the same manner as in (C1) except that (6S) -10-fluoro-1-hydroxy-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (4-c) as starting material instead of (6R) -1- (benzyloxy) -10-fluoro-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Except for triazacyclononane-3-carboxamide (4-a). The product was obtained as TFA salt. MS (m/z): 489.18[ M+H ] ] +1 H NMR (400 MHz, acetonitrile-d 3) delta 10.13 (s, 1H), 8.52 (s, 1H), 7.32 (dd, j=8.4, 5.8hz, 1H), 7.23-7.08 (m, 1H), 7.08-6.97 (m, 1H), 6.88 (t, j=8.5 hz, 2H), 5.56 (d, j=16.7 hz, 1H), 4.83-4.69 (m, 2H), 4.64 (d, j=5.7 hz, 2H), 4.58-4.49 (m, 2H), 4.13 (d, j=13.3 hz, 1H).
Preparation of (6S) -9-fluoro-1-hydroxy-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (C4)
The synthesis of the title product was carried out in the same manner as in (C1) except that (6S) -9-fluoro-1-hydrogen-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,7,12,14-tetrahydro-6, 13-methanobenzo [ g]Pyrido [1,2-b][1,2,5]Triazacyclononane-3-carboxamide (4-d) as starting material instead of (6R) -1- (benzyloxy) -10-fluoro-2, 14-dioxo-N- (2, 4, 6-trifluorobenzyl)-2,7,12,14-tetrahydro-6, 13-methanobenzo [ g ]]Pyrido [1,2-b][1,2,5]Except for triazacyclononane-3-carboxamide (4-a). The product was obtained as TFA salt. MS (m/z): 489.19[ M+H ]] +1 H NMR (400 MHz, acetonitrile-d 3) delta 10.14 (s, 1H), 8.53 (d, j=14.2 hz, 1H), 7.36 (dd, j=8.5, 5.7hz, 1H), 7.28-7.01 (m, 2H), 6.88 (t, j=8.6 hz, 2H), 5.51 (d, j=16.3 hz, 1H), 4.79 (q, j=14.5 hz, 2H), 4.71-4.60 (m, 2H), 4.56 (dd, j=15.0, 10.3hz, 2H), 4.20 (d, j=13.6 hz, 1H).
Example 5: preparation of (10S) -6-hydrogen-10-methyl-5, 8-dioxo-N- [ (2, 4, 6-trifluorophenyl) methyl)]-1, 2, 9-Triazatricyclo [7.4.1.02,7 ]]Tetradec-3,6,11-triene-4-carboxamide (C5):
step 1: synthesis of 1- (allyl (t-butoxycarbonyl) amino) -3- (benzyloxy) -4-oxo-5- ((2, 4, 6-tri) Fluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid methyl ester (5)
To 3-benzyloxy-4-oxo-5- [ (2, 4, 6-trifluorophenyl) methylcarbamoyl]To a suspension of methyl pyran-2-carboxylate (2.0 g,4.47 mmol) in a mixture of MeOH (48.0 mL) and water (8.0 mL) was added tert-butyl N-allyl-N-amino-carbamate (0.77 g,4.47 mmol) and sodium bicarbonate (3.76 g,44.7 mmol). The resulting mixture was stirred at room temperature overnight. Water (15.0 mL) was added to the reaction and the mixture was stirred for 10 min. The suspension was filtered and the filter cake was then partitioned between ethyl acetate and water. The aqueous layer was extracted with EtOAc (×2), the combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated to give the desired product which was used directly in the next step. LCMS-esi+ (m/z): c (C) 30 H 30 F 3 N 3 O 7 H+ calculated theoretical value of (2): 601.20, found: 601.99.
step 2: synthesis of N-allyl-N- [ 3-benzyloxy-2- [ [ (1S) -1-methallyl ]Carbamoyl radicals]-4-oxygen Substituted-5-[ (2, 4, 6-Trifluorophenyl) methylcarbamoyl group]-1-pyridinyl]Tert-butyl carbamate (443-int-2) and 1- (allylamino) -3-benzyloxy-N2- [ (1S) -1-methallyl]-4-oxo-N5- [ (2, 4, 6-trifluorophenyl) methyl ]] Pyridine-2, 5-dicarboxamide (6)
Methyl 1- (allyl (t-butoxycarbonyl) amino) -3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylate (5, 2.3g,3.82 mmol) was dissolved in a mixture of MeOH (24.0 mL), THF (12.0 mL) and water (12.0 mL). To this mixture was added lithium hydroxide monohydrate (1.28 g,30.6 mmol). The resulting mixture was heated to 60 ℃ while stirring for 3 hours. The reaction was cooled to room temperature and concentrated. The residue was diluted with EtOAc, acidified to pH-4 with 1N aqueous HCl, the organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated.
The residue was then dissolved in DCM (17.0 mL) and treated with EDCI.HCl (975 mg,5.11 mmol) followed by HOAt (695 mg,5.11 mmol) and DIEA (1.76 g,13.6 mmol) at room temperature. (2S) -but-3-en-2-amine hydrochloride (315 mg,4.43 mmol) was then added. The newly formed mixture was stirred at room temperature overnight. The reaction was then diluted with DCM, saturated NH 4 Cl, brine, dried over sodium sulfate, filtered and concentrated, the residue was mixed with silica gel, concentrated to dryness and purified by combiflash (24 g silica gel, 0-100% EtOAc/hexanes). The desired fractions were combined and concentrated to give N-allyl-N- [ 3-benzyloxy-2- [ [ (1S) -1-methallyl ]]Carbamoyl radicals]-4-oxo-5- [ (2, 4, 6-trifluorophenyl) methylcarbamoyl]-1-pyridinyl]Tert-butyl carbamate (6) (LCMS-ESI+ (m/z): C 33 H 35 F 3 N 4 O 6 H+ calculated theoretical value of (2): 640.25, found: 641.05 1- (allylamino) -3-benzyloxy-N2- [ (1S) -1-methallyl)]-4-oxo-N5- [ (2, 4, 6-trifluorophenyl) methyl ]]Pyridine-2, 5-dicarboxamide (7) (LCMS-ESI+ (m/z): H+ calculated theory of C28H27F3N4O 4: 540.20, found: 541.02).
Then 6 (1.0 g) at room temperature was treated with 4N HCl in 1, 4-dioxane (10.0 mL,40.0 mmol) at room temperature in DCM (10.0 m)The solution in L) for 1 hour, converting compound 6 into compound 7. The reaction was concentrated and co-evaporated with etoac×3. The residue was then dissolved in MeOH (20 mL) and NaHCO was added 3 (solid), stirred for 15 minutes, filtered, the filtrate concentrated and repurified by combiflash to give compound 7.
Step 3: synthesis of 1-allyl-5-hydroxy-3- [ (1S) -1-methallyl ]4, 6-dioxo-N- [ (2, 4, 6-) Trifluorophenyl) methyl]-2H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide (8) and 1-allyl-5-benzyloxy-3- [ (1S) -1-methylallyl]-4, 6-dioxo-N- [ (2, 4, 6-trifluorophenyl) methyl)]-2H-pyrido [2,1-f][1,2, 4]Triazine-7-carboxamide (9)
1- (allylamino) -3-benzyloxy-N2- [ (1S) -1-methallyl at room temperature]-4-oxo-N5- [ (2, 4, 6-trifluorophenyl) methyl ]]Pyridine-2, 5-dicarboxylic acid amide (7) (350 mg,0.647 mmol) was dissolved in a mixture of ACN (3.5 mL) and DCE (3.5 mL). Paraformaldehyde (58.4 mg,0.647 mmol) was added to the mixture. The resulting mixture was then heated to 88 ℃. To the hot mixture was added dropwise acetic acid (0.35 mL), followed by TFA (0.15 mL). The reaction was capped and heated for an additional 30 minutes. The reaction was cooled to room temperature, diluted with EtOAc, and saturated NaHCO 3 Alkalizing to pH 7, washing the organic layer with brine, drying over sodium sulfate, filtering and concentrating. The residue was purified by combiflash (12 g silica gel, 0-100% EtOAc, dry-loaded) to give the desired 1-allyl-5-benzyloxy-3- [ (1S) -1-methallyl]-4, 6-dioxo-N- [ (2, 4, 6-trifluorophenyl) methyl)]-2H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide (9) LCMS-esi+ (m/z): h+ calculated theory for C29H27F3N4O 4: 552.20, found: 552.93. debenzylated form (8) was also isolated. LCMS-esi+ (m/z): h+ calculated theory for C22H21F3N4O 4: 462.15, found: 463.02.
Compound 8 (120 mg,0.26 mmol) was converted back to compound 9 by treating compound 8 (120 mg,0.26 mmol) in DMF (2.6 mL) with benzyl bromide (46.6 mg,0.272 mmol) and cesium carbonate (101 mg,0.31 mmol) overnight at room temperature. The reaction was diluted with EtOAc, washed with water, brine, dried over sodium sulfate, filtered, mixed with silica gel, concentrated to dryness and purified by combiflash (4 g silica gel, 0-100% EtOAc/hexanes) to give 9.LCMS-esi+ (m/z): h+ calculated theory for C29H27F3N4O 4: 552.20, found: 552.92.
step 4: synthesis of (10S) -6-benzyloxy-10-methyl-5, 8-dioxo-N- [ (2, 4, 6-trifluorophenyl) methyl]- 1,2, 9-triazatricyclo [7.4.1.02,7 ]]Tetradecane-3,6,11-triene-4-carboxamide (10)
Compound 9 (130 mg,0.23 mmol) was dissolved in DCM (29 mL) and Hoveyda-Grubbs II catalyst (36.9 mg,0.059 mmol) was added to the mixture. The resulting mixture was sparged with nitrogen for 5 minutes, then capped and heated at 70 ℃ overnight. The reaction was then cooled to room temperature, concentrated and purified by normal phase chromatography (12 g silica gel, 0-100% etoac/hexanes). LCMS-esi+ (m/z): h+ calculated theory for C27H23F3N4O 4: 524.17, found: 524.91.
step 5: synthesis of (10S) -6-hydrogen-10-methyl-5, 8-dioxo-N- [ (2, 4, 6-trifluorophenyl) methyl ] ]-1,2, 9-triazatricyclo [7.4.1.02,7 ]]Tetradecane-3,6,11-triene-4-carboxamide (C5)
Compound 10 (8 mg,0.015 mmol) was dissolved in DCM (1.0 mL) at room temperature and treated with TFA (1.0 mL) for 3 hours at room temperature. The reaction was concentrated, redissolved in DMF, filtered and purified by reverse phase preparative HPLC. LCMS-esi+ (m/z): h+ calculated theory for C20H17F3N4O 4: 434.12, found: 435.19.1H NMR (400 MHz, DMSO-d 6) δ10.34 (t, J=5.8 Hz, 1H), 8.32 (s, 1H), 7.26-7.17 (m, 2H), 5.75-5.66 (m, 1H), 5.46 (ddt, J=12.0, 6.2,3.3Hz, 1H), 5.34-5.22 (m, 1H), 4.92 (d, J=14.4 Hz, 1H), 4.72 (d, J=14.4 Hz, 1H), 4.65-4.50 (m, 3H), 4.27-4.17 (m, 1H), 3.70-3.62 (m, 1H), 1.29 (d, J=7.3 Hz, 3H).
Example 6: preparation of 13-benzyl-8-hydroxy-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-) Hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (C6)
Step 1: synthesis of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) Methyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid methyl ester
The reactor was charged with tert-butyl N-carbamate (390 mg,2.95 mmol), naHCO in MeOH/water (9 ml/6 ml) 3 (457mg, 5.4 mmol) followed by the addition of methyl 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -4H-pyran-2-carboxylate (1200 mg,2.68 mmol). The reaction mixture was heated to 60 ℃ overnight. The reaction was cooled to room temperature and extracted with ethyl acetate (100 ml). The organic layer was concentrated in vacuo. The residue was used in the next reaction without purification. MS (m/z) 562.064[ M+H ] ] +
Step 2: synthesis of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) Radical) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid
To the above residue in MeOH (6 mL) was added 2.5N LiOH solution (2 mL) at room temperature. After 2 hours at room temperature, the reaction was acidified with 2N HCl and extracted with ethyl acetate (100 ml). The organic layer was concentrated in vacuo. The residue was used in the next reaction without purification. MS (m/z) 547.96[ M+H ]] +
Step 3: synthesis of (3- (benzyloxy) -2-carbamoyl-4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) Phenyl) pyridin-1 (4H) -yl) carbamic acid tert-butyl ester
To a solution of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid (1720 mg,3.14 mmol) in DMF (12 ml) was added EDC (015 mg,6.28 mmol), HOBt (720 mg,4.7 mmol), DIPEA (4060 mg,31.4 mmol) and ammonium chloride (1680 mg,31.4 mmol) at room temperature. After stirring overnight at room temperature, the reaction was diluted with ethyl acetate (100 ml) and washed with brine. The organic layer was dried over MgSO 4 Dried and concentrated in vacuo. The resulting residue was purified by column chromatography. MS (m/z) 547.029[ M+H ]] + 。1H NMR(400MHz, chloroform-d) delta 10.12 (t, j=5.7 Hz, 1H), 8.58 (s, 1H), 8.50 (s, 1H), 7.37 (dq, j=4.1, 3.0,2.4Hz, 5H), 6.70 (dd, j=8.7, 7.5Hz, 3H), 5.84 (s, 1H), 5.33 (d, j=4.5 Hz, 3H), 4.68 (d, j=5.7 Hz, 2H), 1.45 (s, 9H).
Step 4: synthesis of 1-amino-3- (benzyloxy) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) -1, 4-dihydropyridine- 2, 5-dicarboxamides
To a solution of 1-amino-3- (benzyloxy) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) -1, 4-dihydropyridine-2, 5-dicarboxamide (200 mg,0.366 mmol) in DCM (6 ml) was added TFA (0.5 ml) at room temperature after stirring at room temperature for 2 hours, the solvent and excess TFA were removed in vacuo and the resulting residue was used in the next reaction without purification.ms (m/z) 447.075[ m+h ]] +
Step 5: synthesis of 2-benzyl-5- (benzyloxy) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetralin hydrogen-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
To a solution of 1-amino-3- (benzyloxy) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) -1, 4-dihydropyridine-2, 5-dicarboxamide (165 mg,0.37 mmol) in DMF (2 ml) was added AcOH (2 ml) and 2-phenylacetaldehyde (44 mg,0.37 mmol) at room temperature after heating to 100deg.C for 2 hours, DMF and excess AcOH were removed in vacuo and the remaining residue was purified by column chromatography MS (m/z) 549.065[ M+H] +
Step 6: synthesis of 13-benzyl-8- (benzyloxy) -7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
Solid KOH (106 mg,1.9 mmol) was suspended in DMF (12 ml) and 2-benzyl-5- (benzyloxy) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] was added by syringe pump at 0deg.C over 2 hours][1,2,4]A mixed solution of triazine-7-carboxamide (129 mg,0.235 mmol) and 1, 4-dibromobutane (56 mg,0.26 mmol) in DMF (10 ml). The reaction mixture was extracted with ethyl acetate (100 ml). The organic layer was concentrated in vacuo. The residue was used in the next reaction without purification. MS (m/z) 601.033[ M-H] +
Step 7: synthesis of 13-benzyl-8-hydroxy-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexa-form Hydrogen-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (C6)
To crude 13-benzyl-8- (benzyloxy) -7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] at room temperature][1,2,5]To a solution of triazacyclononane-10-carboxamide (129 mg,0.225 mmol) in toluene (5 ml) was added TFA (1 ml). After stirring overnight at room temperature, the solvent and excess TFA were removed. The resulting residue was purified by prep HPLC to give the title compound TFA salt. MS (m/z) 513.253[ M+H ]] +1 H NMR (400 MHz, chloroform-d) δ10.37 (d, j=5.9 hz, 1H), 9.13 (s, 1H), 8.57 (s, 1H), 7.28 (s, 5H), 7.11 (d, j=7.0 hz, 1H), 6.69 (t, j=8.3 hz, 2H), 4.90-4.60 (m, 2H), 4.56-4.22 (m, 2H), 3.58-3.24 (m, 2H), 3.06-2.74 (m, 2H).
Example 7: preparation of (1S, 2R,6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluoro) Benzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene (triazecine) -11- Formamide (C7)
Step 1: synthesis of (R) -3- (benzyloxy) -1- ((tert-butoxycarbonyl) (pent-4-en-2-yl) amino) -4-oxo- 5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid methyl ester (7A)
To a reaction mixture of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid methyl ester (3.5 g,6.23 mmol), (2S) -pent-4-en-2-ol (805 mg,9.35 mmol) and triphenylphosphine (3.27 g,12.5 mmol) in 7mL THF was added diisopropyl azodicarboxylate (2.45 mL,12.5 mmol). The resulting reaction mixture was stirred at room temperature for 30 min and concentrated in vacuo. The residue was purified by silica gel chromatography eluting with EtOAc/hexanes to give the title product. MS (m/z) 630.10[ M+H ] +.
Step 2: synthesis of (R) -3- (benzyloxy) -1- ((tert-butoxycarbonyl) (pent-4-en-2-yl) amino) -4-oxo- 5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid (7B)
(R) -3- (benzyloxy) -1- ((tert-butoxycarbonyl) (pent-4-en-2-yl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid methyl ester (7A, 3.17g,5.04 mmol) was dissolved in MeOH (20 mL), THF (30 mL) and water (10 mL). Lithium hydroxide monohydrate (1.05 g,25.2 mmol) was added. The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with EtOAc, acidified to pH-4 with 1N HCl, the organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated to give the title product. MS (m/z) 616.20[ M+H ] +.
Step 3: synthesis of (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2), 4, 6-Trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) ((R) -pent-4-en-2-yl) carbamic acid tert-butyl ester (7C)
To a reaction mixture of (R) -3- (benzyloxy) -1- ((tert-butoxycarbonyl) (pent-4-en-2-yl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid (7 b,2.1g,3.41 mmol), (2S) -but-3-en-2-amine hydrochloride (477 mg,4.43 mmol), edci.hcl (977 mg,5.12 mmol) and HOAt (696 mg,5.12 mmol) in DCM (34 mL) was added N, N-diisopropylethylamine (2.38 mL,13.6 mmol). The reaction mixture was stirred at room temperature for 30 min, diluted with DCM, and saturated NH 4 Cl and brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography eluting with EtOAc/hexanes to give the title product. MS (m/z) 669.83[ M+H ]]+。
Step 4: synthesis of (2R, 6S, Z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-11- ((2, 4, 6-trifluoro) Benzyl) carbamoyl) -2,3,6,7,8,10-hexahydro-1H-pyrido [1,2-b][1,2,5]Triazene-1-carboxylic acid tert-butyl ester (7D)
A solution of tert-butyl (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) ((R) -pent-4-en-2-yl) carbamate (7C, 1.0g,1.5 mmol) and the 2 nd generation Grubbs catalyst (63.5 mg,0.075 mmol) in 500ml toluene was purged with Ar for 30 minutes. The resulting solution was heated in an oil bath at 80 ℃ for 5 hours. The reaction mixture was concentrated and the residue was purified by column chromatography on silica gel eluting with EtOAc in hexanes to give the title product. MS (m/z) 641.29[ M+H ] +.
Step 5: synthesis of (2R, 6S, Z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,3,6,7,8,10-hexahydro-1H-pyrido [1,2-b][1,2,5]Triazene-11-carboxamide (7E)
(2R, 6S, Z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-11- ((2, 4, 6-trifluorobenzyl) carbamoyl-2,3,6,7,8,10-hexahydro-1H-pyrido [1, 2-b)][1,2,5]Tert-butyl triazene-1-carboxylate (7D, 170mg,0.265 mmol) was dissolved in DCM (5 ml) and treated with 4N HCl in 1, 4-dioxane (3 ml) for 3 hours at room temperature. Then more 4N HCl in 1, 4-dioxane (2 ml) was added and stirred at room temperature for 2 hours. After concentrating to dryness, the residue was dissolved in EtOAc and taken up with saturated NaHCO 3 And brine wash. The organic layer was dried over MgSO 4 Drying, filtration and concentration to dryness followed by high vacuum drying gives the title product. MS (m/z) 541.24[ M+H ]]+。
Step 6: synthesis of (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamides (7F)
(2R, 6S, Z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,6,7,8,10-hexahydro-1H-pyrido [1,2-b ] in an 8mL sample vial ][1,2,5]Triazene-11-carboxamide (7E, 50mg, 0.093) and paraformaldehyde (6.1 mg,2.2 equivalents based on MW: 30) are mixed with acetonitrile (1.25 mL) and DCE (1.25 mL) at room temperature, capped and immediately placed on a hot plate preheated at 88 ℃. Into which to dropAcOH (0.25 mL,10% acetonitrile) was added followed by dropwise TFA (0.25 mL,10% DCE solution). The resulting reaction mixture was then heated for an additional 30 minutes. Cool to room temperature and pour into well-stirred EtOAc-NaHCO 3 (aqueous solution) in a two-phase mixture. The organic phase was separated. The aqueous layer was extracted once with EtOAc. The combined organic phases were washed with brine, dried over Na 2 SO 4 Drying and filtering. The residue was concentrated to dryness and purified by RP-HPLC to give the title product. MS (m/z) 553.17[ M+H ]]+。
Step 7: synthesis of (1S, 2R,6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamide (C7)
(1S, 2R,6S, Z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamide (7F, 30mg,0.054 mmol) was dissolved in 2mL toluene to which was added 2mL TFA. The mixture was stirred at room temperature for 40 minutes. The solvent was removed and the residue was purified by RP-HPLC to give the title product. The structure was confirmed by X-ray crystallography. MS (m/z): 463.20[ M+H ] ]+。 1 H NMR (400 MHz, acetonitrile-d 3) delta 10.42 (s, 1H), 8.42 (s, 1H), 6.94-6.81 (m, 2H), 5.75-5.60 (m, 2H), 5.20 (q, j=7.1, 6.7hz, 1H), 4.82-4.56 (m, 4H), 3.59 (p, j=6.9 hz, 1H), 2.39 (dd, j=15.5, 7.3hz, 1H), 2.06 (ddd, j=16.7, 7.9,5.5hz, 1H), 1.29 (t, j=7.3 hz, 6H).
Example 8: preparation of (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamides (C8)
/>
(1S, 2R,6S, Z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11Formamide (7F, 35mg,0.063 mmol) was dissolved in 3mL ethanol and 3mL EtOAc and sparged under argon. Palladium on carbon (10 wt%, wet) (13.5 mg) was added and the mixture was sparged under a hydrogen atmosphere (1 atm, balloon). The mixture was vigorously stirred for two hours and then sprayed under an argon atmosphere. By passing throughAnd (5) filtering the pad. Washing ∈10 with absolute ethanol>And the filtrate was concentrated to dryness and the residue was purified by RP-HPLC to give the title product. MS (m/z): 465.200[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d 3) delta 10.37 (s, 1H), 8.32 (s, 1H), 6.94-6.81 (m, 2H), 4.87 (d, j=14.3 hz, 1H), 4.71-4.59 (m, 3H), 4.39 (tt, j=11.5, 6.6hz, 1H), 2.95 (dq, j=8.6, 6.3hz, 1H), 2.10-1.97 (m, 2H), 1.87-1.72 (m, 2H), 1.63-1.49 (m, 1H), 1.30 (q, j=12.0 hz, 1H), 1.13 (dd, j=14.3, 6.5hz, 6H).
Example 9: preparation of (1 'S,5' S) -8 '-hydroxy-5, 5' -trimethyl-7 ',9' -dioxo-N- (2, 4, 6-trio) Fluorobenzyl) -4,5,7',9' -tetrahydro-2H, 5 'H-spiro [ furan-3, 2' - [1,6]]Methanopyrido [1,2-b ] bridged][1,2,5] Triazacyclononane]-10' -carboxamide (C9)
(1 ' S,5' S) -8' -hydroxy-5, 5' -trimethyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) -4,5,7',9' -tetrahydro-2H, 5' H-spiro [ furan-3, 2' - [1,6] methanopyrido [1,2-b ] [1,2,5] triazacyclononane ] -10' -carboxamide was prepared in analogy to example 84 except that 5, 5-dimethyltetrahydrofuran-3-carbaldehyde was used instead of tetrahydrofuran-3-carbaldehyde in step 1 and only one product was isolated from the ring closing metathesis reaction in step 9. MS (m/z) 519.24[ M+H ] +.1H NMR (400 MHz, methanol-d 4) delta 8.79 (s, 1H), 6.91 (t, J=8.4 Hz, 2H), 6.01 (dd, J=11.9, 2.3Hz, 1H), 5.49 (dd, J=12.0, 2.8Hz, 1H), 5.19 (d, J=14.7 Hz, 1H), 5.01 (d, J=14.7 Hz, 1H), 4.67 (s, 2H), 4.37 (dt, J=7.5, 2.6Hz, 1H), 3.73-3.61 (m, 2H), 2.52 (dd, J=13.5, 1.6Hz, 1H), 2.07-1.94 (m, 1H), 1.87 (d, J=7.4 Hz, 3H), 1.56 (s, 3H), 1.34 (s, 3H).
Example 10 and example 11: preparation of (1R, 2R, Z) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10- Dioxo-3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ] ][1,2,5]Triazene-11-carboxamide (C10) And (1S, 2R, Z) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7- Methanopyrido [1,2-b ] bridged][1,2,5]Triazene-11-carboxamide (C11)
(2R, Z) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide (9A) was prepared in a similar manner to (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide (7F in example 7), using 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylic acid methyl ester in place of step 1 3- (benzyloxy) -1-tert-butoxycarbonyl) -4-oxo-1, 4-dihydropyridine-2-carboxylic acid methyl ester in place of step 1, and butan-2- ((3, 2,5] triazene-carboxamide (7F in example 7). MS (m/z): 521.20[ M+H ] +.
It was separated into its individual diastereomers (10B and 11A) by preparative SFC chromatography on an IA column using ethanol as a co-solvent. The separated diastereomers were dissolved in 1mL toluene and 1mL TFA and stirred at room temperature for 1 hour. After concentration, purification by RP-HPLC eluting with ACN/water (0.1% TFA) provided the title compounds C10 and C11.
Peak 1: (1R, 2R, Z) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-810-dioxo-3,6,8,10-tetratetralin hydrogen-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C10)):MS(m/z):431.20[M+H]+。 1 H NMR (400 MHz, acetonitrile-d 3) delta 10.37 (s, 1H), 8.35 (s, 1H), 7.44 (d, j=7.4 hz, 1H), 6.96 (d, j=9.8 hz, 2H), 5.83 (q, j=9.7, 8.6hz, 1H), 5.66-5.58 (m, 1H), 5.15 (d, j=13.8 hz, 1H), 4.94 (d, j=17.8 hz, 1H), 4.68-4.57 (m, 3H), 3.55-3.38 (m, 2H), 2.25 (dt, j=16.7, 8.5hz, 1H), 1.97-1.86 (m, 1H), 1.15 (d, j=6.9 hz, 3H).
Peak 2: (1S, 2R, Z) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10-dioxo-3,6,8,10-tetralin hydrogen-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C11)):MS(m/z):431.20[M+H]+。 1 H NMR (400 MHz, acetonitrile-d 3) delta 10.41 (s, 1H), 8.44 (s, 1H), 7.44 (q, j=8.8, 8.3hz, 1H), 6.97 (tt, j=10.8, 3.1hz, 2H), 5.79-5.64 (m, 2H), 4.98-4.88 (m, 2H), 4.69-4.58 (m, 3H), 3.61 (t, j=6.7 hz, 1H), 3.53 (dd, j=18.0, 4.0hz, 1H), 2.42 (dd, j=15.4, 7.1hz, 1H), 2.16-2.03 (m, 1H), 1.29 (d, j=7.1 hz, 3H).
Example 12: preparation of (1R, 2S,6R, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluoro) Benzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamide (C12)
Compound 12 is prepared in analogy to (1S, 2R,6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Preparation of triazene-11-carboxamide (C7 in example 7) using (2R) -pent-4-en-2-ol instead of (2S) -pent-4-en-2-ol in step 1 and (2R) -but-3-en-2-amine hydrochloride instead of (2S) -but-3-en-2-amine hydrochloride in step 3. MS (m/z): 431.20[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d 3) delta 10.43 (s, 1H), 8.42 (s, 1H), 6.94-6.81 (m, 2H), 5.75-5.60 (m, 2H), 5.20 (q, j=7.5 hz, 1H), 4.84-4.53 (m, 4H), 3.58 (q, j=6.9 hz, 1H), 2.39 (dd, j=15.2, 7.2hz, 1H), 2.12–2.00(m,1H),1.29(t,J=7.3Hz,6H)。
Example 13: preparation of (1S, 2R) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10-dioxo-3, 4, 5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C13)
Compound 13 is prepared in analogy to example 8 (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Preparation of triazene-11-carboxamide (C8) using (1S, 2R, Z) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ] ][1,2,5]Triazene-11-carboxamide (11) replaces (2 r,6s, z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamide (7F). MS (m/z): 433.20[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d) 3 )δ10.39(s,1H),8.34(s,1H),7.57–7.20(m,1H),7.03–6.91(m,2H),4.86(d,J=14.3Hz,1H),4.78(d,J=14.6Hz,1H),4.60(d,J=5.6Hz,2H),4.14(dd,J=13.4,5.9Hz,1H),3.39–2.97(m,2H),1.97–1.87(m,2H),1.75(d,J=6.5Hz,1H),1.63(dt,J=16.9,9.1Hz,1H),1.55–1.20(m,2H),1.17(d,J=6.5Hz,3H)。
Example 14: preparation of (1R, 2S, 6R) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamides (C14)
Compound 14 is prepared in analogy to example 8 (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanoPyrido [1,2-b ] yl][1,2,5]Preparation of triazene-11-carboxamide (C8) by means of (1R, 2S,6R, Z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide replaces (2 r,6s, z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamide (7F). MS (m/z): 465.20[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d 3) delta 10.37 (s, 1H), 8.32 (s, 1H), 6.94-6.81 (m, 2H), 4.87 (d, j=14.3 hz, 1H), 4.74-4.58 (m, 3H), 4.39 (ddd, j=11.8, 6.7,4.8hz, 1H), 3.01-2.89 (m, 1H), 2.07-1.97 (m, 1H), 1.87-1.72 (m, 2H), 1.56 (dt, j=16.6, 10.1hz, 1H), 1.30 (q, j=12.0 hz, 1H), 1.13 (dd, j=14.5, 6.5hz, 6H).
Example 15: preparation of (1R, 2R) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10-dioxo-3, 4, 5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C15)
Compound 15 is prepared in analogy to example 8 (1R, 2R, Z) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Preparation of triazene-11-carboxamide (C8) by means of (1R, 2R, Z) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (10) replaces (2 r,6s, z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamide (7F). MS (m/z): 433.20[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d 3) δ11.50 (s, 1H), 10.41 (s, 1H), 8.32 (d, j=16.7 hz, 1H), 7.49-7.38 (m, 1H), 6.97 (ddt, j=13.4, 8.5,3.0hz, 2H), 5.13 (d, j=14.1 hz, 1H), 4.80 (t, j=14.9 hz, 1H), 4.60 (d, j=6.0 hz, 2H), 4.22 (dt, j=13.8, 4.6hz, 1H), 3.57 (p, j=7.2 hz, 1H), 3.02 (ddd, j=14.0, 10.1,4.2hz, 1H), 1.96 (d, j=2.5 hz, 3H), 1.70-1.55%m,1H),1.53–1.16(m,2H),1.13(d,J=7.0Hz,3H)。
Example 16: preparation of (1S, 2R, Z) -9-hydroxy-2-methyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) scheme 3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-11-carboxamide (C16)
(2R, Z) -9- (benzyloxy) -2-methyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide (16A) was prepared in a similar manner to (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide (7F in example 7), using allylamine hydrochloride instead of (2S) -but-3-en-2-amine hydrochloride in step 3. MS (m/z): 539.20[ M+H ] +.
It was separated into its individual diastereomers (peak 1 and peak 2) by RP-HPLC elution with acetonitrile and water (containing 0.1% tfa). Peak 2 (23 mg,0.043 mmol) dissolved in 0.5mL toluene and 0.5mL TFA was taken and stirred at room temperature for 1 hour. After concentration, purification by RP-HPLC eluting with ACN/water (0.1% TFA) provided the title compound. MS (m/z): 449.10[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d) 3 )δ10.41(s,1H),8.42(s,1H),6.87(t,J=8.7Hz,2H),5.72(q,J=11.7,9.1Hz,2H),4.92(d,J=15.7Hz,2H),4.64(d,J=14.6Hz,3H),3.82–3.11(m,2H),2.46–2.35(m,1H),2.08(dt,J=13.7,6.3Hz,1H),1.28(d,J=7.2Hz,3H)。
Example 17: preparation of (1S, 2R) -9-hydroxy-2-methyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3, 4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] ][1,2,5]Triazene-11-carboxamide (C17)
Compound 17 is prepared in analogy to example 8 (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Preparation of triazene-11-carboxamide (C8) using (1S, 2R, Z) -9-hydroxy-2-methyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (16) replaces (2 r,6s, z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamide (7F). MS (m/z): 451.20[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d 3) delta 10.38 (s, 1H), 8.33 (s, 1H), 6.94-6.81 (m, 2H), 4.85 (d, j=14.4 hz, 1H), 4.76 (d, j=14.5 hz, 1H), 4.62 (d, j=5.6 hz, 2H), 4.14 (dt, j=14.0, 7.0hz, 1H), 3.06 (dt, j=13.8, 4.4hz, 2H), 1.93 (d, j=14.4 hz, 3H), 1.75 (t, j=8.3 hz, 1H), 1.63 (dt, j=16.9, 8.9hz, 1H), 1.44 (q, j=10.1 hz, 1H), 1.16 (d, j=6.5 hz, 3H).
Example 18: preparation of (1S, 2R,6R, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluoro) Benzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazacyclononane-11-carboxamide (C18)
/>
(2R, 6R, Z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide (18A) was prepared in a similar manner to (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide (7F in example 7), using (2R) -but-3-en-2-amine hydrochloride instead of (2S) -but-3-en-2-amine hydrochloride in step 3. MS (m/z): 553.30[ M+H ] +.
It was separated into its individual diastereomers (top and bottom spots) by silica gel chromatography eluting with EtOAc/hexanes. Taking outThe lower spot (15 mg,0.027 mmol) dissolved in 0.5mL toluene and 0.5mL TFA was stirred at room temperature for 1 hour. After concentration, purification by RP-HPLC eluting with ACN/water (0.1% TFA) provided the title compound (18). MS (m/z): 463.20[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d) 3 )δ10.38(s,1H),8.34(s,1H),6.94–6.81(m,2H),5.85–5.72(m,1H),5.53(dt,J=11.6,1.6Hz,1H),5.27(d,J=7.9Hz,1H),4.96(d,J=13.7Hz,1H),4.73(d,J=13.7Hz,1H),4.62(d,J=5.7Hz,2H),3.48–3.35(m,1H),2.29–2.15(m,1H),1.96–1.87(m,1H),1.27(d,J=7.4Hz,3H),1.12(d,J=7.0Hz,3H)。
Example 19 and example 20: preparation of (1R, 2S,6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-Trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11- Formamide (C19) and (1S, 2S,6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3, 6,8, 10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C20)
(2S, 6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide (19A) was prepared in a similar manner to (1S, 2R,6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide (7 in example 7), using (2R) -pent-4-en-2-ol instead of (2S) -pent-4-en-2-ol in step 1. MS (m/z): 463.20[ M+H ] +.
It was separated into its individual diastereomers (peak 1 and peak 2) by preparative SFC chromatography on AZ-H columns using methanol as co-solvent.
Peak 1: (1R, 2S,6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3, 6,8, 10-tetrahydro-2H-1, 7-methanopyrido[1,2-b][1,2,5]Triazene-11-carboxamide (19):MS(m/z):463.20[M+H]+。 1 h NMR (400 MHz, acetonitrile-d 3) delta 11.45 (s, 1H), 10.38 (s, 1H), 8.33 (s, 1H), 6.94-6.81 (m, 2H), 5.85-5.72 (m, 1H), 5.53 (dt, j=11.5, 1.6hz, 1H), 5.27 (d, j=8.0 hz, 1H), 4.96 (d, j=13.7 hz, 1H), 4.72 (d, j=13.6 hz, 1H), 4.62 (d, j=5.7 hz, 2H), 3.48-3.35 (m, 1H), 2.29-2.15 (m, 1H), 1.97-1.85 (m, 1H), 1.27 (d, j=7.4 hz, 3H), 1.12 (d, j=7.hz, 3H).
Peak 2: (1S, 2S,6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3, 6,8, 10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (20):MS(m/z):463.20[M+H]+。 1 h NMR (400 MHz, acetonitrile-d 3) delta 10.44 (s, 1H), 8.39 (s, 1H), 6.87 (t, j=8.5 hz, 2H), 5.69 (d, j=12.1 hz, 1H), 5.59 (dd, j=18.9, 9.5hz, 1H), 4.93 (d, j=14.2 hz, 1H), 4.76-4.42 (m, 3H), 4.06 (s, 1H), 3.55 (s, 1H), 2.60 (s, 1H), 2.07 (t, j=7.4 hz, 1H), 1.81 (d, j=7.4 hz, 3H), 1.29 (d, j=7.1 hz, 3H).
Example 21: preparation of (1R, 2S, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamides (C21)
In analogy to (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] of example 8][1,2,5]Preparation of triazene-11-carboxamide (C8) by means of (1R, 2S,6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C19) replaces (2R, 6S, Z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ][1,2,5]Triazene-11-carboxamide (7F). MS (m/z): 465.20[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d 3) δ10.40 (s, 1H), 8.29(s,1H),6.91–6.83(m,2H),4.84(d,J=14.4Hz,1H),4.77(d,J=14.5Hz,1H),4.69–4.51(m,3H),3.57(q,J=7.1Hz,1H),1.96–1.83(m,2H),1.80–1.54(m,3H),1.39(dd,J=13.0,6.2Hz,1H),1.18(dd,J=7.0,2.3Hz,6H)。
Example 22: preparation of (1S, 2R, 6R) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamides (C22)
In analogy to (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] of example 8][1,2,5]Preparation of triazene-11-carboxamide (C8) by means of (1S, 2R,6R, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C18) replaces (2R, 6S, Z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (7F). MS (m/z): 465.20[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d) 3 )δ10.41(s,1H),8.28(s,1H),6.87(t,J=8.5Hz,2H),4.84(d,J=14.4Hz,1H),4.77(d,J=14.4Hz,1H),4.65–4.52(m,3H),3.55(q,J=7.0Hz,1H),1.95–1.84(m,1H),1.79–1.54(m,3H),1.44–1.34(m,1H),1.18(dd,J=6.9,2.5Hz,7H)。
Example 23: preparation of (1S, 2S, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamides (C23)
Chemical combinationSubstance 23 in analogy to example 8 (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] ][1,2,5]Preparation of triazene-11-carboxamide (C8) using (1S, 2S,6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C20) replaces (2R, 6S, Z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (7F). MS (m/z): 465.20[ M+H ]]+。 1 H NMR(400MHz,DMSO-d 6 )δ11.10(s,1H),10.41(t,J=5.8Hz,1H),8.28(s,1H),7.21(t,J=8.6Hz,2H),6.53(s,0H),5.00(d,J=14.4Hz,1H),4.88(d,J=14.6Hz,1H),4.60(dd,J=14.6,6.0Hz,1H),4.51(dd,J=14.6,5.7Hz,1H),3.53–3.50(m,2H),2.11–1.96(m,1H),1.76(q,J=12.1Hz,1H),1.70–1.61(m,2H),1.53(d,J=6.9Hz,5H),1.32(d,J=7.1Hz,3H)。
EXAMPLE 24 preparation of (1S, 2R, 5S) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C24)
Step 1: preparation of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) Methyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid methyl ester
3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -4H-pyran-2-carboxylic acid methyl ester (60.0 g,134mmol,1.00 eq.) with MeOH (300 mL) and H 2 O (60.0 mL) was mixed. BocNHNH addition at room temperature 2 (19.5 g,147mmol,1.10 eq.) and NaHCO 3 (22.5 g,268mmol,10.4mL,2.00 eq.). The reaction mixture was then stirred at 55 ℃ for 16 hours. The reaction mixture was placed under vacuum to remove most of the MeOH. The obtained residue is treated H for the remainder 2 O (200 mL) was diluted and the crude product extracted with EtOAc (1500 mL). The organic layer was washed with brine (500 mL), and dried over Na 2 SO 4 Dried and concentrated in vacuo. The resulting slurry was purified by silica gel chromatography with petroleum ether ethyl acetate=5:1 to give the product. MS (m/z): 562.5[ M+H ]]。
Step 2: preparation of 3- (benzyloxy) -1- (but-3-en-2-yl (tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-Trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid methyl ester
Methyl 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylate (5 g,8.9 mmol) was dissolved in THF (100 mL) at room temperature. The solution was cooled to 0 ℃ under argon. The but-3-en-2-ol (963 mg,13.4 mmol) and Ph were added sequentially 3 P (3.5 g,13.4 mmol). DIAD (2.7 g,13.4 mmol) was then added dropwise over 5 minutes. The resulting reaction was stirred at 0 ℃ for 5 minutes. The cold bath was removed. The reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was concentrated to dryness. The residue was purified on a silica gel column with 0% -100% etoac/Hex to give the product. MS (m/z): 616.0[ M+H ]]。
Step 3: preparation of 3- (benzyloxy) -1- (but-3-en-2-yl (tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid
Methyl 3- (benzyloxy) -1- (but-3-en-2-yl (tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylate (5.4 g,8.9 mmol) was mixed with MeOH (125 mL) and water (100 mL) at room temperature. LiOH (5M aqueous solution) (11 ml) was added. Assembling air coolingThe reaction mixture was heated to 73 ℃ while stirring for 3.5 hours. More LiOH (5M) (2 mL) was added. The reaction mixture was then stirred at 40 ℃ for 17 hours. The reaction mixture was carefully concentrated to remove MeOH. The residue was diluted and rinsed with some water and acidified with 1N HCl to ph=3. EtOAc (200 mL) was added for extraction. The organic phase was separated. The aqueous layer was extracted with more EtOAc (100 mL). The combined organic phases were washed with water and brine, separated and purified over Na 2 SO 4 Drying, filtering and concentrating to obtain the product. MS (m/z): 602.0[ M+H ]]。
Step 4: preparation of (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2), 4, 6-Trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) (but-3-en-2-yl) carbamic acid tert-butyl ester
/>
3- (benzyloxy) -1- (but-3-en-2-yl (tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid (4.75 g,7.9 mmol) was dissolved in DMF (20 mL) at room temperature. DIEA (6.1 g,47.4 mmol) was added. (S) -but-3-en-2-amine hydrochloride (1.27 g,11.8 mmol) and HATU (4.5 g,11.8 mmol) were then added sequentially. The resulting reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was diluted with EtOAc (100 mL) and then NaHCO 3 (saturated aqueous solution, 100 mL) and water (100 mL). The organic phase was separated and washed with water (50 mL) and brine (50 mL). The final organic phase was concentrated to remove the solvent. The residue was purified on a silica gel column with 0% -100% etoac/Hex to give the product. MS (m/z): 655.0[ M+H ]]。
Step 5: preparation of 3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -1- (but-3-en-2-ylamino) -4-oxo substituted-N5- (2, 4, 6-trifluorobenzyl) -1, 4-dihydropyridine-2, 5-dicarboxamides
Tert-butyl (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) (but-3-en-2-yl) carbamate (4.42 g,6.75 mmol) was dissolved in DCM (10 mL) at room temperature. HCl (4M dioxane solution) (10 mL) was added. The reaction mixture was stirred at room temperature for 6 hours. The reaction mixture was then concentrated to dryness. The residue was partitioned between EtOAc (100 mL) and NaHCO 3 (saturated aqueous solution, 100 mL). The organic phase was separated, washed with brine and over Na 2 SO 4 And (5) drying. The solvent was removed to give the product. MS (m/z): 555.3[ M+H ]]。
Step 6: preparation of 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -1- (but-3-en-2-yl) -4, 6-dioxo substituted-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ][1,2,4]Triazine-7-carboxamide
3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -1- (but-3-en-2-ylamino) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) -1, 4-dihydropyridine-2, 5-dicarboxamide (1.845 g,3.33 mmol) was dissolved in acetonitrile (18.45 mL) and dichloroethane (18.45 mL) at room temperature. Paraformaldehyde (200 mg,6.66 mmol) was added. The resulting mixture was placed on a preheated hot bath at 88 ℃. AcOH (0.9 mL) and TFA (0.9 mL) were then added sequentially to the pre-heated reaction over 5 minutes. The resulting reaction mixture was then sealed and heated with stirring for 30 minutes. The resulting reaction mixture was then concentrated to dryness to remove all solvent and acid. The resulting crude material was then dissolved in DMF (17 mL). Sequentially adding K 2 CO 3 (2.76 g,20 mmol) and benzyl bromide (2.56 g,15 mmol). The reaction mixture was then heated at 100 ℃ for 3 hours. The reaction mixture was then diluted with EtOAc (100 mL) followed by NaHCO 3 (saturated aqueous solution) (100 mL) and water (100 mL). The organic phase was separated and washed with water (50 mL) and brine (50 mL). The solvent was removed in vacuo. Will remain behindThe crude product was purified on a silica gel column with 0% -100% etoac/Hex to give the product. MS (m/z): 567.2[ M+H ]]。
Step 7: preparation of (1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (A), (1S, 2S, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro- 1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (B) and (1R, 2S, 5S) -8- (benzyloxy) Phenyl) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C)
5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -1- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (931 mg,1.64 mmol) was dissolved in dichloroethane (88 mL) at room temperature. Argon was bubbled through the reaction solution for 5 minutes. HG-M720 catalyst (103.4 mg,0.164 mmol) was then added with stirring. Purging with argon was continued for 10 minutes. The reaction mixture was then heated under argon atmosphere with stirring for 48 hours. The resulting reaction mixture was then concentrated to dryness. The crude material was purified on a silica gel column with 0% -100% etoac/Hex to give three diastereomers which could be separated: (1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-B ] [1,2,5] triazacyclononane-10-carboxamide (A), 334mg.MS (m/z) [ 539.2 M+H ] and (1S, 2S, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-B ] [1,2,5] triazacyclononane-10-carboxamide (B), 96mg.MS (m/z) [ 539.2+M+H ] and (1R, 5S) -8- (benzyloxy) -2, 5-dimethyl-N- (2, 4, 6-trifluorobenzyl) -2, 5-tetrahydro-1, 6-methano [1,2,5] triazacyclononane-10-carboxamide (B), 96mg.MS (m/z) [ 539.2 M+H ] and (1S, 2S, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluoromethylpyridinium) -2, 6-methano [1,2, 5-methano-5 ] triazacyclononane-10-carboxamide (B. The absolute configuration of these two compounds remains to be determined.
Preparation of (1S, 2R, 5S) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (C24)
(1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] at room temperature][1,2,5]Triazacyclononane-10-carboxamide (110 mg,0.188 mmol) was dissolved in toluene (1 mL). TFA (1 mL) was carefully added with stirring. The resulting reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was then concentrated to dryness. The residue was dissolved in MeOH and taken up in 0.1% TFA in 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilization gives the product as a mono TFA salt. MS (m/z): 449.2[ M+H ]].1H NMR (400 MHz, acetonitrile-d 3) δ10.21 (t, J=5.9 Hz, 1H), 8.38 (s, 1H), 6.87 (t, J=8.5 Hz, 2H), 5.65 (dt, J=11.4, 2.4Hz, 1H), 5.47-5.27 (m, 2H), 5.01 (d, J=14.4 Hz, 1H), 4.62 (d, J=5.8 Hz, 2H), 4.57 (d, J=14.3 Hz, 1H), 3.82 (tp, J=6.6, 3.3Hz, 1H), 1.35 (d, J=2.0 Hz, 3H), 1.33 (d, J=2.6 Hz, 3H).
Example 25: preparation of (1S, 2S, 5S) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C25)
(1S, 2S, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-)Trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (78 mg,0.145 mmol) was dissolved in toluene (1 mL). TFA (1 mL) was carefully added with stirring. The resulting reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was then concentrated to dryness. The residue was dissolved in MeOH and taken up in 0.1% TFA in 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilization gives the product as a mono TFA salt. 34mg.MS (m/z): 449.2[ M+H ]].1H NMR (400 MHz, acetonitrile-d 3) δ10.28 (s, 1H), 8.44 (s, 1H), 7.11-6.61 (m, 2H), 5.61 (ddd, J=12.3, 3.3,2.1Hz, 1H), 5.50-5.27 (m, 2H), 4.93 (d, J=14.4 Hz, 1H), 4.72 (d, J=14.4 Hz, 1H), 4.66-4.53 (m, 3H), 1.35 (d, J=7.2 Hz, 3H), 1.01 (d, J=7.4 Hz, 3H).
Example 26: preparation of (1R, 2S, 5S) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C26)
(1R, 2S, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (16 mg, mmol) was dissolved in toluene (1 mL) at room temperature. TFA (1 mL) was carefully added with stirring. The resulting reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was then concentrated to dryness. The residue was purified with 0% -100% etoac in hexanes to give the product in neutral form. MS (m/z): 449.2[ M+H ].1H NMR (400 MHz, acetonitrile-d 3) delta 10.30 (s, 1H), 8.40 (s, 1H), 6.97-6.77 (m, 2H), 5.80 (ddd, J=11.7, 2.7,1.9Hz, 1H), 5.45 (ddd, J=11.7, 4.2,2.4Hz, 1H), 4.81-4.69 (m, 2H), 4.65-4.59 (m, 2H), 4.32 (dtt, J=7.5, 5.0,2.5Hz, 1H), 4.01 (ddq, J=7.0, 4.8,2.4Hz, 1H), 1.79 (d, J=7.5 Hz, 3H), 1.40 (d, J=7.0 Hz, 3H).
Example 27: preparation of (1S, 2R, 5S) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluoro)Benzyl Phenyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C27)
(1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (92.8 mg,0.172 mmol) was dissolved in MeOH (10 mL). Pd-C (10%) (23 mg) was added. With H at room temperature 2 The balloon was subjected to hydrogenolysis for 7 hours. The reaction mixture was filtered through celite. The filtrate was collected and concentrated to dryness. The residue was dissolved in MeOH and taken up in 0.1% TFA in 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilization gives the product as a mono TFA salt. MS (m/z): 451.3[ M+H ]].1H NMR (400 MHz, acetonitrile-d 3) delta 10.37 (s, 1H), 8.43 (s, 1H), 7.02-6.78 (m, 2H), 4.71-4.45 (m, 5H), 3.51 (dq, J=7.2, 3.6Hz, 1H), 2.02 (ddd, J=8.2, 6.1,3.3Hz, 1H), 1.78-1.66 (m, 1H), 1.55 (dt, J=8.6, 3.3Hz, 2H), 1.31 (d, J=7.2 Hz, 3H), 1.27 (d, J=6.8 Hz, 3H).
Example 28: preparation of (1S, 2S, 5S) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10-carboxamide (C28)
(1S, 2S, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (18 mg,0.172 mmol) was dissolved in MeOH (8 mL). Pd-C (10%) (10 mg) was added. With H at room temperature 2 The balloon was subjected to hydrogenolysis for 7 hours. The reaction mixture was filtered through celite. Collecting the filtrateConcentrating to dryness. The residue was dissolved in MeOH and taken up in 0.1% TFA in 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilization gives the product as a mono TFA salt. MS (m/z): 451.3[ M+H ]].1H NMR (400 MHz, acetonitrile-d 3) delta 10.36 (s, 1H), 8.34 (s, 1H), 6.98-6.77 (m, 2H), 4.76-4.59 (m, 5H), 3.41 (dd, J=11.6, 6.3Hz, 1H), 2.20 (dt, J=14.2, 6.8Hz, 1H), 1.80-1.49 (m, 2H), 1.26 (d, J=6.7 Hz, 3H), 1.30-1.17 (m, 1H), 1.08 (d, J=6.9 Hz, 3H).
Example 29: preparation of (1S, 2R, 5S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo Substituted-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C29)
Step 1: preparation of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -5- ((2, 4-difluorobenzyl) carbamoyl) Methyl-4-oxo-1, 4-dihydropyridine-2-carboxylate
3- (benzyloxy) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxo-4H-pyran-2-carboxylic acid methyl ester (15 g,34.9 mmol) with MeOH (100 mL) and H 2 O (30.0 mL) was mixed. BocNHNH addition at room temperature 2 (5 g,37.8 mmol) and NaHCO 3 (5.87 g,69.9 mmol). The reaction mixture was then stirred at 55 ℃ for 16 hours. The reaction mixture was placed under vacuum to remove most of the MeOH. The residue obtained is treated with H 2 O (200 mL) was diluted and the crude product extracted with EtOAc (500 mL). The organic layer was washed with brine (500 mL), and dried over Na 2 SO 4 Dried and concentrated in vacuo. The resulting slurry was purified by silica gel chromatography with hexane: ethyl acetate=5:1 to give 18g of the product as a white solid. MS (m/z): 543.95[ M+H ]]。
Step 2: preparation of 3- (benzyloxy) -1- (but-3-en-2-yl (tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4-difluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid methyl ester
Methyl 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4-difluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylate (10.5 g,19.3 mmol) was dissolved in THF (200 mL) at room temperature. The solution was cooled to 0 ℃ under argon. The but-3-en-2-ol (2.37 g,32.8 mmol) and Ph were added sequentially 3 P (8.6 g,32.8 mmol). DIAD (6.64 g,32.8 mmol) was then added dropwise over 5 minutes. The resulting reaction mixture was a slightly orange solution. It was stirred at 0℃for 5 minutes. The cold bath was removed. The reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was concentrated to dryness. The residue was purified on a silica gel column with 0% -100% etoac/Hex to give 10g of product. MS (m/z): 598.04[ M+H ]]。
Step 3: preparation of 3- (benzyloxy) -1- (but-3-en-2-yl (tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4-difluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid
3- (benzyloxy) -1- (but-3-en-2-yl (tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4-difluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid methyl ester (12.5 g,20.9 mmol) was mixed with MeOH (200 mL) and water (100 mL) at room temperature. LiOH (5M in water) (33.5 mL,167 mmol) was added. The reaction mixture was heated to 63 ℃ with stirring for 17 hours, equipped with an air condenser. The reaction mixture was carefully concentrated to remove MeOH. The residue was diluted and rinsed with some water and acidified with 1N HCl to ph=3. Solids appear. EtOAc (200 mL) was added for extraction. The organic phase was separated. The aqueous layer was extracted with more EtOAc (100 mL). The combined organic phases were washed with water and brine. The organic phase was taken up in Na 2 SO 4 Drying, filtration and concentration gave 9g of the acidic product. MS (m/z): 584.30[ M+H ]]。
Step 4: preparation of (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2), 4-difluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) (but-3-en-2-yl) carbamic acid tert-butyl ester
3- (benzyloxy) -1- (but-3-en-2-yl (tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid (5 g,8.56 mmol) was dissolved in DMF (40 mL) at room temperature. DIEA (5.52 g,42.8 mmol) and the reaction mixture were cooled with an ice-water bath. HATU (6.52 g,17.14 mmol) was added in one portion. The reaction mixture was then warmed to room temperature and stirred for 1 hour. (S) -but-3-en-2-amine hydrochloride (2.3 g,21.4 mmol) was then added. The resulting reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was diluted with EtOAc (200 mL) and then NaHCO 3 (saturated aqueous solution, 100 mL) and water (100 mL). The organic phase was separated and washed with water (50 mL) and brine (50 mL). The final organic phase was concentrated to remove the solvent. The residue was sufficiently pure to be used in the next step. 4g. MS (m/z): 637.03[ M+H ]]。
Step 5: preparation of 3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -1- (but-3-en-2-ylamino) -4-oxo substituted-N5- (2, 4-difluorobenzyl) -1, 4-dihydropyridine-2, 5-dicarboxamide
Tert-butyl (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2, 4-difluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) (but-3-en-2-yl) carbamate (4 g,6.29 mmol) was dissolved in DCM (10 mL) at room temperature. HCl (4M dioxane solution) (10 mL) was added. The reaction mixture was stirred at room temperature for 6 hours. The reaction mixture was then concentrated to dryness. The residue was partitioned between EtOAc (100 mL) and NaHCO 3 (saturated aqueous solution, 100 mL). The organic phase was separated, washed with brine and over Na 2 SO 4 And (5) drying. The solvent was removed to give 3g of a product. MS (m/z): 537.17[ M+H ]]。
Step 6: preparation of 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -1- (but-3-en-2-yl) -4, 6-dioxo substituted-N- (2, 4-difluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -1- (but-3-en-2-ylamino) -4-oxo-N5- (2, 4-difluorobenzyl) -1, 4-dihydropyridine-2, 5-dicarboxamide (2.52 g,4.7 mmol) was dissolved in acetonitrile (25 mL) and dichloroethane (25 mL) at room temperature. Paraformaldehyde (278 mg,9.25 mmol) is added. The resulting slurry was placed on a preheated hot bath at 88 ℃. AcOH (1.15 mL) and TFA (1.15 mL) were then added sequentially to the pre-heated reaction over 5 minutes. The resulting reaction mixture was then sealed and heated with stirring for 30 minutes. The resulting reaction mixture was then concentrated to dryness to remove all solvent and acid. The resulting crude material was then dissolved in DMF (17 mL). Sequentially adding K 2 CO 3 (10 g,72.4 mmol) and benzyl bromide (6.63 g,38.8 mmol). The reaction mixture was then heated at 100 ℃ for 3 hours. The reaction mixture was then diluted with EtOAc (100 mL) followed by NaHCO 3 (saturated aqueous solution) (100 mL) and water (100 mL). The organic phase was separated and washed with water (50 mL) and brine (50 mL). The solvent was removed in vacuo. The residual crude product was purified on a silica gel column with 0% -100% etoac/Hex to give 1g of product. MS (m/z): 549.2[ M+H ]]。
Step 7: preparation of (1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4-difluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide and (1S, 2S, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4-difluorobenzyl) -2,5,7, 9-tetrahydro-1,6-Bridge Methylene pyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide
5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -1- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4-difluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (753 mg,1.37 mmol) was dissolved in dichloroethane (83 mL) at room temperature. Argon was bubbled through the reaction solution for 5 minutes. HG-M720 catalyst (146.4 mg,0.233 mmol) was then added with stirring. Purging with argon was continued for 10 minutes. The reaction mixture was then heated under argon atmosphere with stirring for 48 hours. The resulting reaction mixture was then concentrated to dryness. The crude material was purified on a silica gel column with 0% -100% etoac/Hex to give two products as single diastereomers: (1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4-difluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide 230mg.MS (m/z): 521.2[ M+H ] and (1S, 2S, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4-difluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide 15mg.MS (m/z): 521.2 M+H ]. The absolute configuration of these two compounds remains to be determined.
Step 8: preparation of (1S, 2R, 5S) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4-difluorobenzyl) -2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] at room temperature][1,2,5]Triazacyclononane-10-carboxamide (230 mg,0.188 mmol) was dissolved in toluene (1 mL). TFA (1 mL) was carefully added with stirring. Will be spentThe reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was then concentrated to dryness. The residue was dissolved in MeOH and taken up in 0.1% TFA in 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilization gives the product as a mono TFA salt. MS (m/z): 431.2[ M+H ]].1H NMR (400 MHz, acetonitrile-d 3) δ10.21 (s, 1H), 8.39 (s, 1H), 7.44 (td, J=8.8, 6.5Hz, 1H), 7.21-6.84 (m, 2H), 5.66 (dt, J=11.4, 2.4Hz, 1H), 5.46-5.32 (m, 2H), 5.02 (d, J=14.4 Hz, 1H), 4.67-4.50 (m, 3H), 3.84 (qq, J=6.6, 3.0Hz, 1H), 1.35 (dd, J=7.1, 4.1Hz, 6H).
Example 30: preparation of (1S, 2S, 5S) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4-difluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10-carboxamide (C30)
(1S, 2S, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4-difluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] at room temperature][1,2,5]Triazacyclononane-10-carboxamide (15 mg,0.0288 mmol) was dissolved in toluene (1 mL). TFA (1 mL) was carefully added with stirring. The resulting reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was then concentrated to dryness. The residue was dissolved in MeOH and taken up in 0.1% TFA in 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilization gives the product as a mono TFA salt. MS (m/z): 431.2[ M+H ]].1H NMR (400 MHz, acetonitrile-d 3) δ10.27 (s, 1H), 8.45 (s, 1H), 7.44 (td, J=8.8, 6.5Hz, 1H), 6.97 (dddd, J=10.7, 5.2,4.3,2.5Hz, 2H), 5.62 (ddd, J=12.2, 3.3,2.1Hz, 1H), 5.46-5.31 (m, 2H), 4.94 (d, J=14.4 Hz, 1H), 4.73 (d, J=14.4 Hz, 1H), 4.61 (d, J=6.1 Hz, 3H), 1.36 (d, J=7.2 Hz, 3H), 1.03 (d, J=7.4 Hz, 3H).
Example 31: preparation of (1S, 2S, 5S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo Substituted-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triaza-sCyclononane-10-carboxamide (C31)
/>
(1S, 2S, 5S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ][1,2,5]A solution of triazacyclononane-10-carboxamide (example 30,0.29 mmol,125 mg) in EtOH (9 mL) was evacuated and backfilled with argon (5 Xcycle), then treated with 10% Pd/C (25 mg), and further evacuated and backfilled with argon (5 Xcycle), then with hydrogen (5 Xcycle). The reaction mixture was stirred at room temperature under a hydrogen balloon overnight, then filtered through celite, washed with EtOH and concentrated. The resulting residue was redissolved in EtOH (22 mL) and treated with 10% Pd/C (50 mg) and hydrogen as described above. After 4 hours, the reaction mixture was filtered through celite, washed with EtOH and concentrated, then purified by preparative HPLC (10% -100% mecn in water with 0.1% tfa) and lyophilized to give the title compound as trifluoroacetate salt (62 mg,39% yield). MS (m/z) 433.25[ M+H ]]+。 1 H NMR (400 MHz, methanol-d 4) delta 8.43 (s, 1H), 7.43 (td, j=8.5, 6.4hz, 1H), 7.02-6.88 (m, 2H), 4.85-4.76 (m, 2H), 4.69 (dt, j=10.6, 6.7hz, 1H), 4.63 (s, 2H), 3.55-3.44 (m, 1H), 2.22 (dt, j=14.2, 6.8hz, 1H), 1.75 (dt, j=15.0, 11.3hz, 1H), 1.66 (dd, j=15.7, 6.9hz, 1H), 1.30 (d, j=6.7 hz, 3H), 1.36-1.20 (m, 1H), 1.11 (d, j=6.8 hz, 3H).
Example 32: preparation of (1S, 2R,3R,4S, 5S) -8-hydroxy-3, 4-dimethoxy-2, 5-dimethyl-7, 9-dioxo substituted-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triaza-s Cyclononane-10-carboxamide: (C32)
Preparation of (1S, 2R,3R,4S, 5S) -8- (benzyloxy) -3, 4-dihydroxy-2, 5-dimethylRadical-7, 9-dioxo-N-) (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclonones Alkane-10-carboxamides
(1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] in a 50mL flask][1,2,5]Triazacyclononane-10-carboxamide (example 24, step 7, A) (140 mg,0.26 mmol) was dissolved in a mixture of acetone (8 mL) and water (1 mL) and then cooled to 0deg.C in an ice-water bath. 4-methylmorpholine N-oxide (50% aqueous solution, 0.1mL,2 eq.) was slowly added to the reaction solution. Then 2.5% osmium tetroxide (0.01 mL,4% equiv.) was added. The mixture was stirred at 0 ℃ and warmed to room temperature for 2 days. The reaction was quenched by addition of 10% aqueous sodium sulfite solution. The reaction mixture was extracted with (1:1) EtOAc: n-BnOH. The organic layer was concentrated and purified by silica column (eluting with 0-10% meoh/DCM) to give (1 s,2r,3s,4r,5 s) -8- (benzyloxy) -3, 4-dihydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1, 2-b) ][1,2,5]Triazacyclononane-10-carboxamide (nonpolar, secondary product) and (1S, 2R,3R,4S, 5S) -8- (benzyloxy) -3, 4-dihydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (polar, major product). MS (m/z) 573.3[ M+H] +
Preparation of (1S, 2R,3R,4S, 5S) -8- (benzyloxy) -3, 4-dimethoxy-2, 5-dimethyl-7, 9-dioxo-N-) (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclonones Alkane-10-carboxamide, (1S, 2R,3R,4S, 5S) -8- (benzyloxy) -3-hydroxy-4-methoxy-2, 5-dimethyl-7, 9-dioxo substituted-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]TrinitrogenImpurity(s) Cyclononane-10-carboxamide and (1S, 2R,3R,4S, 5S) -8- (benzyloxy) -4-hydroxy-3-methoxy-2, 5-dimethyl-7, 9- Dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Three kinds of Azacyclononane-10-carboxamide
(1S, 2R,3R,4S, 5S) -8- (benzyloxy) -3, 4-dihydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] in a 50mL flask][1,2,5]Triazacyclononane-10-carboxamide (77 mg,0.134 mmol) was dissolved in DMF (3 mL) and then cooled to 0deg.C in an ice-water bath. Sodium hydride (60%, 11mg,2.2 eq) was added to the above reaction solution. After 10 minutes, methyl iodide (100-fold dilution with DCM, 1mL,1.3 eq.) was added. The mixture was stirred at 0 ℃ for 30 minutes. LC-MS shows a bis-Me product and two mono-Me products and some starting materials. The reaction was quenched by addition of saturated aqueous sodium bicarbonate. The reaction mixture was extracted with ethyl acetate. The organic layer was concentrated and purified by preparative HPLC eluting with 10% to 100% acetonitrile (0.1% tfa) in water (0.1% tfa) to give (1 s,2r,3r,4s,5 s) -8- (benzyloxy) -3, 4-dimethoxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] ][1,2,5]Triazacyclononane-10-carboxamide MS (m/z) 601.3[ M+H] + ) And (1S, 2R,3R,4S, 5S) -8- (benzyloxy) -3-hydroxy-4-methoxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (polar, predominantly mono-Me product MS (m/z) 587.3[ M+H)] + ) And (1S, 2R,3R,4S, 5S) -8- (benzyloxy) -4-hydroxy-3-methoxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (polar, minor mono-Me product MS (m/z)) 587.3[ M+H] + )
Preparation of (1S, 2R,3R,4S, 5S) -8-hydroxy-3, 4-dimethoxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide [ ]C32):
(1S, 2R,3R,4S, 5S) -8- (benzyloxy) -3, 4-dimethoxy-2, 5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (15 mg) was dissolved in toluene (0.5 mL), and trifluoroacetic acid (1 mL) was added. The reaction was stirred at room temperature for 2 hours. The reaction was concentrated and purified via preparative HPLC eluting with 10% to 100% acetonitrile (0.1% tfa) in water (0.1% tfa) to give 7.2mg of the title compound. MS (m/z) 511.3[ M+H ] ] + .1H NMR (400 MHz, acetonitrile-d 3) delta 10.40 (s, 1H), 8.45 (s, 1H), 6.99-6.80 (m, 2H), 4.70-4.59 (m, 2H), 4.52 (t, J=14.8 Hz, 2H), 4.26-4.12 (m, 1H), 3.81 (d, J=7.6 Hz, 1H), 3.61 (d, J=3.1 Hz, 1H), 3.47 (d, J=8.8 Hz, 1H), 3.36 (s, 3H), 3.09 (s, 3H), 1.48-1.25 (m, 6H).
Example 33: preparation of (1S, 2R,4R, 5S) -8-hydroxy-4-methoxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclonones Alkane-10-carboxamide (C33)
Preparation of (1S, 2R,4R, 5S) -8- (benzyloxy) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trio Fluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxylic acid Amine and (1S, 2R,3S, 5S) -8- (benzyloxy) -3-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) o-N- 2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl was placed in a 100mL flask1, 2-b-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2- ]][1,2,5]Triazacyclononane-10-carboxamide (example 24, step 7, A) (384 mg,0.71 mmol) was dissolved in isopropanol (15 mL) and purged with argon. Phenylsilane (2 eq) and Shenvi catalyst (tris [ (Z) -1-tert-butyl-4, 4-dimethyl-3-oxo-pent-1-enoxy) ]Manganese) (3%) was added to the above reaction solution. An oxygen balloon is used. The mixture was stirred at room temperature for one day. LC-MS shows the product, still raw material. The reaction was quenched by addition of 10% aqueous sodium thiosulfate. The reaction mixture was extracted with ethyl acetate. The organic layer was concentrated and purified via silica column (eluting with 0% -10% meoh/DCM) to give (1 s,2r,4r,5 s) -8- (benzyloxy) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1, 2-b)][1,2,5]Triazacyclononane-10-carboxamide (polar product, MS (m/z) 573.3[ M+H ]] + NMR HMBC, COSY, NOE study confirmed the structure) and (1S, 2R,3S, 5S) -8- (benzyloxy) -3-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (nonpolar, MS (m/z) MS (m/z) 573.3[ M+H)] + )。
Preparation of (1S, 2R,4R, 5S) -4, 8-dihydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R,4R, 5S) -8- (benzyloxy) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10-carboxamide (4 mg) was dissolved in toluene (0.2 mL), and trifluoroethylene was addedAcid (0.4 mL). The reaction was stirred at room temperature for 2 hours. The reaction was concentrated and purified via preparative HPLC eluting with 10% -100% acetonitrile (0.1% tfa) in water (0.1% tfa) to give the title compound. MS (m/z) 467.2[ M+H] + .1H NMR (400 MHz, acetonitrile-d 3) delta 10.35 (s, 1H), 8.40 (s, 1H), 6.87 (t, J=8.5 Hz, 2H), 4.71-4.43 (m, 4H), 4.36 (p, J=6.8 Hz, 1H), 3.88 (dd, J=9.2, 6.0Hz, 1H), 3.49 (m, 1H), 1.87-1.69 (m, 1H), 1.51-1.19 (m, 7H).
Preparation of (1S, 2R,4R, 5S) -8- (benzyloxy) -4-methoxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-) Trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-A Amides and their use
(1S, 2R,4R, 5S) -8- (benzyloxy) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] in a 50mL flask][1,2,5]Triazacyclononane-10-carboxamide (10 mg,0.018 mmol) was dissolved in DMF (1 mL) and then cooled to 0deg.C in an ice-water bath. Sodium hydride (60%, 1.4mg,2 eq) was added to the above reaction solution. After 10 minutes, methyl iodide (100-fold dilution with DCM, 0.11mL,1 eq) was added. The mixture was stirred at 0 ℃ for 30 minutes. LC-MS shows a bis-Me product, mainly a mono-Me product. The reaction was quenched by addition of saturated aqueous sodium bicarbonate. The reaction mixture was extracted with ethyl acetate. The organic layer was concentrated and purified via preparative HPLC eluting with 10% -100% acetonitrile (0.1% tfa) in water (0.1% tfa) to give the title compound. MS (m/z) 571.3[ M+H ] +
Preparation of (1S, 2R,4R, 5S) -8-hydroxy-4-methoxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluoro) Benzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C33)
(1S, 2R,4R, 5S) -8- (benzyloxy) -4-hydroxy1, 2-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (4 mg) was dissolved in toluene (0.2 mL), and trifluoroacetic acid (0.4 mL) was added. The reaction was stirred at room temperature for 2 hours. The reaction was concentrated and purified via preparative HPLC eluting with 10% to 100% acetonitrile (0.1% tfa) in water (0.1% tfa) to give 1.6mg of the title compound. MS (m/z) 481.2[ M+H ]] +1 H NMR (400 MHz, acetonitrile-d 3) delta 10.33 (s, 1H), 8.42 (d, j=8.2 hz, 1H), 6.87 (t, j=8.5 hz, 2H), 4.76-4.46 (m, 4H), 3.45 (dd, j=9.0, 5.4hz, 2H), 3.30 (s, 3H), 1.90-1.74 (m, 2H), 1.55-1.21 (m, 7H).
Example 34: preparation of (1S, 2R,4S, 5S) -4-fluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10- Formamide (C34)
Preparation of (1S, 2R,4S, 5S) -8- (benzyloxy) -4-fluoro-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluoro) Benzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxylic acid Amines
(1S, 2R,4R, 5S) -8- (benzyloxy) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] in a 50mL flask][1,2,5]Triazacyclononane-10-carboxamide (28 mg,0.05 mmol) was dissolved in DCM (1.5 mL) and then cooled to 0deg.C in an ice-water bath. Bis (2-methoxyethyl) aminothiotrifluoride (50% toluene solution, 2.7M,0.037mL,2 eq.) was added to the above reaction solution. The mixture was stirred at 0 ℃ for 30 minutes. LC-MS showed fluorinated products and elimination products. The reaction was quenched by addition of saturated aqueous sodium bicarbonate. The reaction mixture was extracted with ethyl acetate. The organic layer was concentrated, attempted purification via normal phase silica column and preparative HPLC,without separation. The mixture was dissolved in a mixture of acetone (4 mL) and water (0.5 mL) and cooled to 0 ℃ in an ice-water bath. 4-methylmorpholine N-oxide (50% in water, 0.017mL,1.5 eq.) was slowly added to the reaction solution. Then 2.5% osmium tetroxide (0.023 mL,4% equiv.) was added. The mixture was stirred at 0 ℃ and warmed to room temperature for 2 days. The reaction was quenched by addition of 10% aqueous sodium sulfite solution. The reaction mixture was extracted with ethyl acetate. The organic layer was concentrated and purified by preparative HPLC eluting with 10% to 100% acetonitrile (0.1% tfa) in water (0.1% tfa) to collect the non-polar product to give (1 s,2r,4s,5 s) -8- (benzyloxy) -4-fluoro-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1, 2-b) ][1,2,5]Triazacyclononane-10-carboxamide (nonpolar). MS (m/z) 559.3[ M+H ]] +
Preparation of (1S, 2R,4S, 5S) -4-fluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C34)
(1S, 2R,4S, 5S) -8- (benzyloxy) -4-fluoro-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (8 mg) was dissolved in toluene (0.2 mL), and trifluoroacetic acid (0.4 mL) was added. The reaction was stirred at room temperature for 2 hours. The reaction was concentrated and purified via preparative HPLC eluting with 10% -100% acetonitrile (0.1% tfa) in water (0.1% tfa) to give the title compound. MS (m/z) 469.2[ M+H] +1 H NMR (400 MHz, acetonitrile-d 3) delta 10.26 (s, 1H), 8.41 (s, 1H), 7.01-6.75 (m, 2H), 5.11-4.92 (m, 1H), 4.91-4.81 (m, 2H), 4.66-4.59 (m, 2H), 3.49-3.34 (m, 1H), 2.39-2.23 (m, 1H), 1.95-1.79 (m, 1H), 1.51,1.30 (m, 1H), 1.45 (dd, J=7.1, 2.0Hz, 3H), 1.35 (dd, J=7.2, 2.9Hz, 3H).
Example 35: preparation (1)S,2R, 5S) -4, 4-difluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10- Formamide: (C35)
Preparation of (1S, 2R, 5S) -8- (benzyloxy) -2, 5-difluoro-4, 7, 9-trioxo-N- (2, 4, 6-trifluorobenzyl) -2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R,4R, 5S) -8- (benzyloxy) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] in a 50mL flask][1,2,5]Triazacyclononane-10-carboxamide (127 mg,0.228 mmol) was dissolved in DCM (3 mL) and then cooled to 0deg.C in an ice-water bath. Dess-martin periodate (194 mg,2 eq) was added to the above reaction solution. The mixture was stirred at 0 ℃ and warmed to room temperature for 2 hours. The reaction was quenched by addition of 10% aqueous sodium thiosulfate. The reaction mixture was extracted with ethyl acetate. The organic layer was concentrated and purified via silica column (eluting with 0% -10% meoh/DCM) to give (1 s,2r,5 s) -8- (benzyloxy) -2, 5-dimethyl-4, 7, 9-trioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1, 2-b)][1,2,5]Triazacyclononane-10-carboxamide (93 mg). MS (m/z) 555.3[ M+H ]] +
Preparation of (1S, 2R, 5S) -8- (benzyloxy) -4, 4-difluoro-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trio) Fluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxylic acid Amines
(1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-4, 7, 9-trioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] in a 50mL flask][1,2,5]Triazacyclononane-10-carboxamide (110 mg,0.2 mmol) was dissolvedIn DCM (5 mL) and then cooled to 0deg.C in an ice-water bath. Bis (2-methoxyethyl) aminothiotrifluoride (50% toluene solution, 2.7M,0.147mL,2 eq.) was added to the above reaction solution. The mixture was stirred at 0 ℃ and warmed to room temperature overnight. The reaction was quenched by addition of saturated aqueous sodium bicarbonate. The reaction mixture was extracted with ethyl acetate. The organic layer was concentrated and purified via silica column (eluting with 0% -10% meoh/DCM) to give (1 s,2r,5 s) -8- (benzyloxy) -4, 4-difluoro-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1, 2-b)][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 577.2[ M+H ]] +
Preparation of (1S, 2R, 5S) -4, 4-difluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10-carboxamide (C35)
(1S, 2R, 5S) -8- (benzyloxy) -4, 4-difluoro-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (25 mg) was dissolved in toluene (0.5 mL) and trifluoroacetic acid (2 mL) was added. The reaction was stirred at room temperature for 2 hours. The reaction was concentrated and purified via preparative HPLC eluting with 10% -100% acetonitrile (0.1% tfa) in water (0.1% tfa) to give the title compound. MS (m/z) 487.2[ M+H ]] +1 H NMR(400MHz,DMSO-d6)δ10.47(s,1H),10.29(t,J=5.8Hz,1H),8.24(s,1H),7.31–7.16(m,2H),5.76(s,1H),5.07(d,J=15.2Hz,1H),4.91(dq,J=15.0,7.2Hz,1H),4.76(d,J=15.2Hz,1H),4.57(d,J=5.8Hz,2H),3.21(t,J=8.3Hz,1H),2.83(ddd,J=32.9,16.3,10.1Hz,2H),1.33(dd,J=7.0,3.1Hz,6H)。
Example 36: preparation of (1S, 2R, 5S) -3-fluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trio) Fluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide: (C36)
preparation of (1S, 2R, 5S) -8- (benzyloxy) -2, 5-difluoro-3, 7, 9-trioxo-N- (2, 4, 6-trifluorobenzyl) -2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R,3S, 5S) -8- (benzyloxy) -3-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] in a 25mL flask][1,2,5]Triazacyclononane-10-carboxamide (16 mg,0.028 mmol) was dissolved in DCM (1 mL) and then cooled to 0deg.C in an ice-water bath. Dess-martin periodate (11 mg,2 eq) was added to the above reaction solution. The mixture was stirred at 0 ℃ and warmed to room temperature for 2 hours. The reaction was quenched by addition of 10% aqueous sodium thiosulfate. The reaction mixture was extracted with ethyl acetate. The organic layer was concentrated and purified via silica column (eluting with 0% -10% meoh/DCM) to give (1 s,2r,5 s) -8- (benzyloxy) -2, 5-dimethyl-3, 7, 9-trioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1, 2-b) ][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 555.3[ M+H ]] +
Preparation of (1S, 2R, 5S) -8- (benzyloxy) -3-fluoro-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-3, 7, 9-trioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] in a 50mL flask][1,2,5]Triazacyclononane-10-carboxamide (12 mg,0.2 mmol) was dissolved in DCM (2 mL) and then cooled to 0deg.C in an ice-water bath. Bis (2-methoxyethyl) aminothiotrifluoride (50% toluene solution, 2.7M,0.018mL,2 eq.) was added to the above reaction solution. The mixture was stirred at 0 ℃ and warmed to room temperature for 3 hours. LC-MS showed elimination product and di-F product. The reaction was quenched by addition of saturated aqueous sodium bicarbonate. Extraction of the reaction mixture using ethyl acetateAnd (3) a compound. The organic layer was concentrated and purified by preparative HPLC eluting with 10% to 100% acetonitrile (0.1% tfa) in water (0.1% tfa) to give (1 s,2r,5 s) -8- (benzyloxy) -3-fluoro-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1, 2-b) ][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 557.3[ M+H ]] +
Preparation of (1S, 2R, 5S) -3-fluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) scheme 2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C36)
(1S, 2R, 5S) -8- (benzyloxy) -3-fluoro-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (6 mg) was dissolved in toluene (0.2 mL) and trifluoroacetic acid (0.5 mL) was added. The reaction was stirred at room temperature for 2 hours. The reaction was concentrated and purified via preparative HPLC eluting with 10% -100% acetonitrile (0.1% tfa) in water (0.1% tfa) to give the title compound. MS (m/z) 467.2[ M+H] +1 H NMR (400 MHz, acetonitrile-d 3) delta 10.20 (s, 1H), 8.43 (s, 1H), 6.87 (t, j=8.5 hz, 2H), 5.70-5.48 (m, 1H), 5.32-5.17 (m, 1H), 5.00 (d, j=14.8 hz, 1H), 4.78-4.57 (m, 3H), 4.22-4.08 (m, 1H), 1.52 (dd, j=6.9, 3.0hz, 3H), 1.37 (d, j=7.2 hz, 3H).
Example 37: preparation of (1S, 2R, 5S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo Substituted-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (C37)
To (1S, 2R, 5S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ][1,2,5]To a solution of triazacyclononane-10-carboxamide (10.2 mg,0.023 mmol) in EtOH (5 mL) was added PtO 2 (2 mg,0.009 mmol). The reaction is carried outAt room temperature at H 2 Stirred under balloon for 2 hours. The reaction mixture was filtered through celite, the filtrate was concentrated and the residue was purified by reverse phase HPLC eluting with 5% -100% aqueous acetonitrile to give the desired product. MS (m/z): 433.11[ M+H ]] +1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 7.51-7.38 (m, 1H), 6.97 (q, j=10.0, 9.5hz, 2H), 4.78 (d, j=14.3 hz, 1H), 4.65 (s, 4H), 3.57 (s, 1H), 2.05 (dd, j=15.0, 7.6hz, 1H), 1.82 (dd, j=15.1, 9.4hz, 1H), 1.62 (d, j=12.2 hz, 2H), 1.40 (d, j=7.0 hz, 3H), 1.33 (d, j=6.7 hz, 3H).
Example 38: preparation of (1S, 2R,4R, 5S) -N- (2, 4-difluorobenzyl) -4, 5-difluoro-9-hydroxy-2-methyl-8, 10-dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxylic acid Amine (C38)
Step 1: synthesis of (1S, 2R,4S, 5R) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 5-dihydroxy-2-methyl 1, 2-b-methyl-8, 10-dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazenes- 11-carboxamide (15A)
(1S, 2R, Z) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ] in 3mL of acetone and 0.45mL of water ][1,2,5]Triazene-11-carboxamide (11A, 190mg,0.36 mmol) was cooled to 0 ℃. To this was added 4-methylmorpholine n-oxide (50% aqueous solution, 0.076ml,0.36 mmol) and osmium tetroxide (2.5% t-BuOH solution, 0.15ml,0.0014 mmol). The reaction was stirred at room temperature for 2 days. To the reaction was added 10% aqueous sodium sulfite solution (3 mL) and stirred for 15 minutes. It was extracted three times with ethyl acetate. The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated to dryness. The residue was purified by silica gel chromatography eluting with methanol in dichloromethane to give the title product. The two hydroxyl stereogenic centers are arbitrarily specified. MS (m/z) 555.300[M+H] +
Step 2: synthesis of (1S, 2R,4R, 5S) -N- (2, 4-difluorobenzyl) -4, 5-difluoro-9-hydroxy-2-methyl-8, 10-) Dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamides (C38)
(1S, 2R,4S, 5R) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 5-dihydroxy-2-methyl-8, 10-dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] in dichloromethane (2 mL)][1,2,5]Triazene-11-carboxamide (10 mg,0.018 mmol) was cooled to 0℃under argon. To this was added Deoxo-fluoro (50% toluene solution, 0.02ml,0.054 mmol). The resulting mixture was stirred at 0 ℃ for three hours. The reaction mixture was diluted with dichloromethane, cooled in an ice/water bath and quenched by dropwise addition of saturated aqueous sodium bicarbonate. The resulting mixture was stirred for 20 minutes and more saturated aqueous sodium bicarbonate was added until no more bubbling occurred. The organic layer was separated, dried over magnesium sulfate, and the solvent was removed under reduced pressure. The residue was purified by RP-HPLC eluting with acetonitrile/water (containing 0.1% TFA) to give (1S, 2R,4R, 5S) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 5-difluoro-2-methyl-8, 10-dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] ][1,2,5]Triazene-11-carboxamide was dissolved in 0.5ml toluene and 0.5ml TFA. The reaction was stirred at room temperature for one hour. The solvent was removed under reduced pressure and the residue was purified by RP-HPLC eluting with acetonitrile/water (containing 0.1% tfa) to give the title product. MS (m/z) 469.200[ M+H] +1 H NMR (400 MHz, acetonitrile-d 3) delta 10.32 (s, 1H), 8.33 (s, 1H), 7.43 (q, j=9.1, 8.4hz, 1H), 7.00-6.95 (m, 2H), 4.82 (s, 2H), 4.69 (s, 0H), 4.60 (d, j=6.0 hz, 2H), 4.37 (dq, j=8.5, 4.4hz, 1H), 4.19 (dd, j=14.2, 7.9hz, 1H), 3.45 (t, j=7.3 hz, 1H), 3.39-3.31 (m, 1H), 2.29-2.06 (m, 2H), 1.20 (d, j=6.5 hz, 3H).
Example 39: preparation of (1S, 2R,4S, 5R) -N- (2, 4-difluorobenzyl) -9-hydroxy-4, 5-dimethoxy-2-a 1, 2-b-methyl-8, 10-dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazenes- 11-carboxamide (C39)
Step 1: synthesis of (1S, 2R,4S, 5R) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -5-hydroxy-4-methoxy- 2-methyl-8, 10-dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Trinitrogen Alkene-11-carboxamide and (1S, 2R,4S, 5R) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 5-dimethoxy-2-methyl- 8, 10-dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] ][1,2,5]Triazene-11-carboxylic acid methyl ester Amides and their use
(1S, 2R,4S, 5R) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 5-dihydroxy-2-methyl-8, 10-dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide (40 mg,0.072 mmol) was dissolved in 2ml anhydrous DMF and cooled to 0℃using an ice water bath. Sodium hydride (3.8 mg, 60 wt% in oil, 0.094 mmol) was added and the mixture stirred at 0deg.C for 30 min. Methyl iodide (0.0056 ml,0.09 mmol) was added thereto, the mixture was stirred at 0 ℃ for 10 minutes, the reaction was quenched with saturated aqueous ammonium chloride solution and extracted three times with ethyl acetate. The combined organic extracts were washed with 5% aqueous lithium chloride and brine, dried over magnesium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography eluting with ethyl acetate in hexane to give the title product.
(1S, 2R,4S, 5R) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -5-hydroxy-4-methoxy-2-methyl-8, 10-dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide: (m/z) 569.300[ M+H ] +.
(1S, 2R,4S, 5R) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 5-dimethoxy-2-methyl-8, 10-dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide: (m/z) 583.300[ M+H ] +.
Step 2: synthesis of (1S, 2R,4S, 5R) -N- (2, 4-difluorobenzyl) -9-hydroxy-4, 5-di-Methoxy-2-methyl-) 8, 10-dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxylic acid methyl ester Amides (C39)):
(1S, 2R,4S, 5R) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 5-dimethoxy-2-methyl-8, 10-dioxo-3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamide (10 mg,0.017 mmol) was dissolved in 0.5ml toluene and 0.5ml TFA and stirred at room temperature for 20 minutes. The solvent was removed under reduced pressure and the residue was purified by RP-HPLC eluting with acetonitrile/water (containing 0.1% tfa) to give the title product. MS (m/z) 493.200[ M+H ]] +1 H NMR (400 MHz, acetonitrile-d) 3 )δ10.37(s,1H),8.32(s,1H),7.49–7.38(m,1H),6.97(ddt,J=14.4,8.4,3.0Hz,2H),4.81(d,J=15.0Hz,1H),4.59(d,J=6.2Hz,3H),3.79(dd,J=7.0,3.6Hz,2H),3.46–3.37(m,0H),3.41(s,3H),3.33(s,3H),3.32(d,J=13.2Hz,1H),3.19(d,J=33.1Hz,1H),3.20-3.08(m,1H),2.18(s,1H),1.94(d,J=2.5Hz,0H),1.89–1.74(m,0H),1.18(d,J=6.5Hz,3H)。
Example 40: preparation of (1S, 2S) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10-dioxo-3, 4, 5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C40)
Step 1: synthesis of tert-butyl (tert-butoxycarbonyl) (prop-2-yn-1-yl) carbamic acid tert-butyl ester
To a stirred solution of prop-2-yn-1-amine (100 g,181.65 mmol) in acetonitrile (2L) was added di-tert-butyl dicarbonate (991 g,4541 mmol) in portions at room temperature followed by DMAP (221.9 g,181.65 mmol) and then stirred at room temperature for 4 hours. After the reaction was completed, the reaction mixture was quenched with water and quenched with acetic acid Ethyl ester extraction. The combined organic layers were washed with brine, dried over Na 2 SO 4 Drying and evaporation under reduced pressure gave a crude compound, which was purified by silica gel column chromatography, eluting with ethyl acetate in hexane to give the title product as a colourless liquid. MS (m/z): 256.31[ M+H ]]+。
Step 2: synthesis of tert-butyl (R, Z) - (tert-butoxycarbonyl) (5-hydroxyhex-2-en-1-yl) carbamate
To a stirred solution of tert-butyl (tert-butoxycarbonyl) (prop-2-yn-1-yl) carbamate (100 g,390.6 mmol) in THF (1L) was added dropwise n-BuLi (1.6M in hexane, 244.14ml,390.6 mmol) at-78deg.C for 30 min. Adding BF thereto 3 Etherate (119.7 g,390.6 mmol) was then added dropwise to a solution of (R) -2-methyl oxirane (21.51 g,390.6 mmol) in THF (0.5L). The reaction mixture was stirred at this temperature for 4 hours. After completion, the reaction mixture was quenched with saturated ammonium chloride solution and water. The reaction mixture was extracted with ethyl acetate. The combined organic layers were washed with saturated NaHCO 3 Washing with solution and brine, passing through Na 2 SO 4 Dried and evaporated to obtain a crude product, which was purified by silica gel column chromatography eluting with a hexane solution of ethyl acetate to obtain tert-butyl (R) - (tert-butoxycarbonyl) (5-hydroxyhex-2-yn-1-yl) carbamate as a colorless liquid. 20g (63.89 mmol) was taken, charged to a Parr apparatus, and 400ml of ethyl acetate was added. Lindlar catalyst (4.4 g) was added under an inert atmosphere followed by quinoline and hydrogenated at 30Psi for 16 hours at room temperature. After completion, the reaction mixture was filtered through a celite pad. The celite pad was washed with ethyl acetate. The filtrate was washed with 1N HCl and concentrated under reduced pressure to obtain a crude product, which was purified by silica gel column chromatography eluting with ethyl acetate in hexane to obtain the title product. MS (m/z): 316.29[ M+H ] ]+。
Step 3: synthesis of (R, Z) -6-amino hex-4-en-2-ol hydrochloride
A solution of tert-butyl (R, Z) - (tert-butoxycarbonyl) (5-hydroxyhex-2-en-1-yl) carbamate (14 g,44.44 mmol) and 4M HCl in dioxane (210 mL) was stirred at room temperature for 1 hour. After completion, the reaction mixture was concentrated under reduced pressure and the crude product was washed with diethyl ether, filtered and dried in vacuo to give the title product. MS (m/z): 116.3[ M+H ] +.
Step 4: synthesis of tert-butyl (R, Z) - (5-hydroxyhex-2-en-1-yl) carbamate
To a stirred solution of (R, Z) -6-amino hex-4-en-2-ol hydrochloride (2 g,13.2 mmol) in 30ml DCM was added di-tert-butyl dicarbonate (4.3 g,19.8 mmol) followed by triethylamine (5.52 ml,39.6 mmol) at room temperature. The reaction mixture was stirred overnight and concentrated to dryness. The crude compound was purified by silica gel column chromatography eluting with ethyl acetate in hexane to give the title product. 1 H NMR (400 MHz, chloroform-d) delta 5.59 (dq, j=10.4, 5.4hz, 2H), 4.67 (s, 1H), 3.94-3.78 (m, 2H), 3.71 (dd, j=15.0, 5.0hz, 1H), 2.44-2.18 (m, 2H), 1.46 (s, 9H), 1.25 (d, j=6.3 hz, 3H).
Step 5: synthesis of (S, Z) -3- (benzyloxy) -1- ((tert-butoxycarbonyl) (6- ((tert-butoxycarbonyl) amino) Hex-4-en-2-yl) amino) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylic acid methyl ester Esters (26E)
Prepared in a similar manner to (R) -methyl 3- (benzyloxy) -1- ((tert-butoxycarbonyl) (pent-4-en-2-yl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylate (7A), using methyl 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylate and tert-butyl (R, Z) - (5-hydroxyhex-2-en-1-yl) carbamate in place of methyl 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylate and (2S) -pent-4-en-2-ol. MS (m/z): 741.400[ M+H ] +.
Step 6: synthesis of (S, Z) -1- ((6-aminohex-4-en-2-yl) amino) -3- (benzyloxy) -5- ((2, 4-difluoro) Benzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylic acid hydrochloride (26F)
To (S, Z) -3- (benzyloxy) -1- ((tert-butoxycarbonyl) (6- ((tert-butoxycarbonyl) amino) hex-4-ene-2-yl) amino) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylic acid methyl ester (5.4 g,7.29 mmol) in 140ml THF/MeOH/H 2 Lithium hydroxide (698 mg,29.2 mmol) was added to the solution in O (3/2/1). The reaction mixture was stirred at 60 ℃ for 2 hours. LCMS showed high conversion to formic acid. The reaction mixture was diluted with ethyl acetate and acidified to pH-4 with 1N HCl. The organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated to give (S, Z) -3- (benzyloxy) -1- ((tert-butoxycarbonyl) (6- ((tert-butoxycarbonyl) amino) hex-4-en-2-yl) amino) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylic acid, which was dissolved in 50mL DCM, treated with 4N HCl in 1, 4-dioxane (7.3 mL) overnight at room temperature and concentrated to dryness. Drying under high vacuum gives the title product. MS (m/z): 527.300[ M+H ]]+。
Step 7: synthesis of (S, Z) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-methyl-8, 10-dioxo-2, 3,6, 7,8, 10-hexahydro-1H-pyrido [1,2-b][1,2,5]Triazene-11-carboxamides
(S, Z) -1- ((6-amino-4-en-2-yl) amino) -3- (benzyloxy) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylate (5.29 g,8.8 mmol) was dissolved in 500ml DCM. EDCI. HCl (2.5 g,13.2 mmol), HOAt (1.8 g,13.2 mmol) and then N, N-diisopropylethylamine (7.69 ml,44.1 mmol) were added thereto. The reaction mixture was stirred at room temperature for 10 min, washed with water and brine. The organic layer was separated, dried over magnesium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography eluting with EtOAc/hexanes to give the title product. MS (m/z): 509.300[ M+H ] +.
Step 8: synthesizing (1R, 2S, Z) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-methyl-8, 10-dioxo-3, 6,8, 10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (26H) and (1 s,2s, z) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7-methano Pyrido [1,2-b ] yl][1,2,5]Triazene-11-carboxamides
Prepared in a similar manner to (1S, 2r, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) 3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide (7F) using (S, Z) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-methyl-8, 10-dioxo-2,3,6,7,8,10-hexahydro-1H-pyrido [1,2-b ] [1,2,5] triazene-11-carboxamide instead of (2 r,6S, Z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,6,7,8,10-hexahydro-1H-pyrido [1,2-b ] [1,2,5] triazene-11-carboxamide (7E). After silica gel chromatography, elution with ethyl acetate in hexane afforded two products:
peak 1: (1 r,2s, z) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide: MS (m/z): 521.300[ M+H ] +.
Peak 2: (1 s,2s, z) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ] [1,2,5] triazene-11-carboxamide: MS (m/z): 521.300[ M+H ] +.
Step 9: synthesis of (1S, 2S) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10-dioxo-3, 4,5,6, 8, 10-hexahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C40)
In analogy to (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Preparation of triazene-11-carboxamide (C8) using (1S, 2S, Z) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide replaces (2 r,6s, z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) 3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamide (7F). MS (m/z): 433.200[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d) 3 )δ11.45(s,1H),10.40(s,1H),8.29(s,1H),7.49–7.38(m,1H),7.04–6.91(m,2H),5.12(d,J=14.1Hz,1H),4.80(d,J=14.2Hz,1H),4.59(d,J=5.9Hz,2H),4.21(dt,J=13.8,4.7Hz,1H),3.57(p,J=7.2Hz,1H),3.02(ddd,J=14.1,10.1,4.2Hz,1H),1.97(p,J=2.5Hz,3H),1.84–1.70(m,1H),1.71–1.54(m,2H),1.50–1.25(m,1H),1.12(d,J=7.0Hz,3H)。
Example 41: preparation of (1S, 2S, Z) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10-dioxo-3, 6,8, 10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C41)
In analogy to (2R, 6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Preparation of triazene-11-carboxamide (C7) by means of (1S, 2S, Z) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide replaces (1 s,2r,6s, z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) 3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (7F). MS (m/z): 431.200[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d) 3 )δ10.37(s,1H),8.35(s,1H),7.50–7.39(m,1H),6.97(ddt,J=12.9,8.4,3.0Hz,2H),5.89–5.77(m,1H),5.62(ddt,J=11.8,4.3,1.8Hz,1H),5.14(d,J=13.8Hz,1H),4.94(d,J=18.1Hz,1H),4.68–4.57(m,3H),3.55–3.38(m,2H),2.26(dt,J=16.1,8.4Hz,1H),1.97–1.86(m,1H),1.15(d,J=7.0Hz,3H)。
Example 42: preparing (1R, 2S) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10-dioxo-3, 4, 5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C42)
In analogy to (1S, 2R, 6S) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,4,5,6,8,10-hexahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Preparation of triazene-11-carboxamide (C8) by means of (1R, 2S, Z) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide replaces (2 r,6s, z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) 3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ][1,2,5]Triazene-11-carboxamide (7F). MS (m/z): 433.200[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d 3) delta 10.38 (s, 1H), 8.35 (s, 1H), 7.44 (td, j=9.2, 8.8,6.5hz, 1H), 7.03-6.91 (m, 2H), 4.86 (d, j=14.5 hz, 1H), 4.78 (d, j=14.5 hz, 1H), 4.60 (d, j=5.8 hz, 2H), 4.15 (ddd, j=14.1, 7.4,5.2hz, 1H), 3.14-3.02 (m, 2H), 1.96-1.91 (m, 1H), 1.77 (dd, j=16.7, 7.1hz, 1H), 1.71-1.57 (m, 1H), 1.45 (q, j=10.3 hz, 2H), 1.17 (d, j=6.5 hz, 3H).
Example 43: preparing (1R, 2S, Z) -N- (2, 4-difluorobenzyl) -9-hydroxy-2-methyl-8, 10-dioxo-3, 6,8, 10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide (C43)
In analogy to (2R, 6S, Z) -9-hydroxy-2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) -3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Preparation of triazene-11-carboxamide (C7) by means of (1R, 2S, Z) -9- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-methyl-8, 10-dioxo-3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b ]][1,2,5]Triazene-11-carboxamide replaces (2 r,6s, z) -9- (benzyloxy) -2, 6-dimethyl-8, 10-dioxo-N- (2, 4, 6-trifluorobenzyl) 3,6,8,10-tetrahydro-2H-1, 7-methanopyrido [1,2-b][1,2,5]Triazene-11-carboxamide (7F). MS (m/z): 431.200[ M+H ] ]+。 1 H NMR (400 MHz, acetonitrile-d) 3 )δ10.41(s,1H),8.44(s,1H),7.44(td,J=8.8,6.5Hz,1H),6.97(ddt,J=10.8,8.2,3.0Hz,2H),5.79–5.63(m,2H),5.00–4.88(m,2H),4.69–4.57(m,3H),3.61(p,J=6.8Hz,1H),3.58–3.48(m,1H),2.42(dd,J=15.5,7.1Hz,1H),2.10(ddd,J=14.7,8.4,5.5Hz,1H),1.29(d,J=7.1Hz,3H)。
Step 44: preparation of (2S, 5S) -N- (2, 4-difluorobenzyl) -2- (fluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo Substituted-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C44)
Synthesis of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -5- ((2, 4-difluorobenzyl) carbamoyl) -4 ] Oxo-1, 4-dihydropyridine-2-carboxylic acid
Prepared in analogy to 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid (12) in example 6 using methyl 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylate instead of methyl 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylate. MS (m/z): 530.200[ M+H ]] +
Synthesis of (S) - (3- (benzyloxy) -2- (but-3-en-2-ylcarbamoyl) -5- ((2, 4-difluorobenzyl) amino) Formyl) -4-oxopyridin-1 (4H) -yl) carbamic acid tert-butyl ester
A reaction mixture of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylic acid (200 mg,0.378 mmol), (2S) -but-3-en-2-amine hydrochloride (61 mg,0.567 mmol), HATU (172 mg, 0.457 mmol) and HOAt (61.7 mg, 0.457 mmol) in 10ml DMF was cooled to 0 ℃. DIPEA (0.2 ml,1.13 mmol) was added dropwise. The reaction mixture was stirred for 10 minutes and then poured into water. It was extracted three times with ethyl acetate. The combined organic layers were washed with 5% aqueous lithium chloride and brine. The organic layer was dried over MgSO 4 Dried and concentrated in vacuo. The residue was purified by silica gel column chromatography. MS (m/z) 583.300[ M+H ]]+。
Synthesis of (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -5- ((2, 4-difluorobenzyl) ammonia Carbamoyl) -4-oxopyridin-1 (4H) -yl) ((S) -1-hydroxybut-3-en-2-yl) carbamic acid tert-butyl ester
Tert-butyl (S) - (3- (benzyloxy) -2- (but-3-en-2-ylcarbamoyl) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxopyridin-1 (4H) -yl) carbamate (430 mg, 0.488 mmol) and tetrabutylammonium bromide (470 mg,1.48 mmol) were charged to a round bottom flask followed by acetonitrile (20 ml, degassed by argon bubbling immediately before use) followed by the addition of (R, R) -DACH naphthalene Trost ligand (58.4 mg,0.0738 mmol) and tris (dibenzylideneacetone) dipalladium-chloroform adduct (22.9 mg,0.022 mmol). The solution was stirred at room temperature under argon for 20 minutes. Butadiene monoxide (0.149 ml,1.85 mmol) was added. The reaction mixture was stirred at room temperature for 2 hours. The reaction mixture was concentrated in vacuo and purified by column chromatography eluting with ethyl acetate in hexane to give the title product. MS (m/z) 653.300[ M+H] +
Synthesis of (S) -2- ((3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -5- ((2, 4-difluoro) Benzyl) carbamoyl) -4-oxopyridin-1 (4H) -yl) (tert-butoxycarbonyl) amino) but-3-en-1-yl acetate
(3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxopyridin-1 (4H) -yl) ((S) -1-hydroxybut-3-en-2-yl) carbamic acid tert-butyl ester (280 mg,0.429 mmol) was dissolved in 14ml pyridine. To this was added DMAP (105 mg,0.858 mmol) and acetic anhydride (0.40 ml,4.29 mmol). The reaction was heated to 60 ℃ for 2 days, cooled to room temperature and concentrated to dryness. The reaction mixture was partitioned between ethyl acetate and 1N HCl, the organic phase was washed with more 1N HCl, then with saturated NaHCO 3 Aqueous solution and brine wash. The organic phase was dried over MgSO 4 Dried, filtered and concentrated in vacuo. Purification by silica gel column chromatography eluting with ethyl acetate in hexane afforded the title product. MS (m/z) 695.400[ M+H ]] +
Synthesis of (S) -2- ((3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -5- ((2, 4-difluoro) Benzyl) carbamoyl) -4-oxopyridin-1 (4H) -yl) amino) but-3-en-1-yl acetate
(S) -2- ((3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxopyridin-1 (4H) -yl) (tert-butoxycarbonyl) amino) but-3-en-1-yl acetate (202 mg,0.29 mmol) was dissolved in 4ml DCM to which was added hydrogen chloride, 4.0M in 1, 4-dioxane (0.73 ml,2.9 mmol). It was stirred at room temperature for 2 hours. The reaction mixture was concentrated in vacuo and dried under high vacuum to give the title product. . MS (m/z) 595.300[ M+H ] ] +
Synthesis of (S) -2-5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -7- ((2, 4-difluorobenzyl) carbamoyl Phenyl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazin-1-yl) but-3-en-1-yl acetic acid Esters of
In analogy to 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -1- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide was prepared using (S) -2- ((3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxopyridin-1 (4H) -yl) amino) but-3-en-1-yl acetate instead of 3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -1- (but-3-en-2-ylamino) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) -1, 4-dihydropyridine-2, 5-dicarboxamide. MS (m/z) 607.300[ M+H ]] +
Synthesis of ((2S, 5S) -8- (benzyloxy) -10- ((2, 4-difluorobenzyl) carbamoyl) -5-methyl-7, 9-dioxo Substituted-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononan-2-yl) acetic acid methyl ester
In analogy to example 24 (1R, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Preparation of triazacyclononane-10-carboxamide using (S) -2- (5- (benzyloxy) -3 - ((S) -but-3-en-2-yl) -7- ((2, 4-difluorobenzyl) carbamoyl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazin-1-yl) but-3-en-1-yl acetate instead of 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -1- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide. MS (m/z) 579.300[ M+H ]] +
Synthesis of (2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo- 2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide
((2S, 5S) -8- (benzyloxy) -10- ((2, 4-difluorobenzyl) carbamoyl) -5-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1, 2-b)][1,2,5]Methyl triazacyclononan-2-yl) acetate (46 mg,0.79 mmol) was dissolved in 2ml methanol. To this was added potassium carbonate (22 mg,0.16 mmol). The reaction was stirred at room temperature for 10 minutes and then partitioned between ethyl acetate and water. The organic layer was separated, washed with brine, and dried over MgSO 4 Drying and concentrating to dryness gave the title product. MS (m/z) 537.300[ M+H] +
Synthesis of (2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (fluoromethyl) -5-methyl-7, 9-dioxo- 2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10-carboxamide
(2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] in DCM (2 mL)][1,2,5]Triazacyclononane-10-carboxamide (xxH, 15mg,0.028 mmol) was cooled at 0deg.C under argon. To this was added Deoxo-fluoro (50% toluene solution, 0.031ml,0.084 mmol). The resulting mixture was stirred at 0 ℃ for 2 hours. The reaction mixture was diluted with DCM, cooled in an ice/water bath and saturated NaHCO was added dropwise 3 Quenching with water solution. The resulting mixture was stirred for 20 minutes and more saturated NaHCO was added 3 The aqueous solution was stirred for 10 minutes until no more bubbling occurred. The organic layer was separated over Na 2 SO 4 Drying and decompressingAnd (5) removing the solvent. The residue was purified by silica gel chromatography eluting with EtOAc/hexanes to give the title product. MS (m/z) 539.200[ M+H ]] +
Synthesis of (2S, 5S) -N- (2, 4-difluorobenzyl) -2- (fluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-2, 5, 7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C44)
(2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (fluoromethyl) -5-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10-carboxamide (11 mg,0.02 mmol) was dissolved in 0.5ml toluene and 0.5ml TFA and stirred at room temperature for 3 hours. The solvent was removed under reduced pressure and the residue was purified by RP-HPLC eluting with acetonitrile/water (containing 0.1% tfa) to give the title product. MS (m/z) 449.200[ M+H ]] +1 H NMR (400 MHz, acetonitrile-d) 3 )δ10.19(s,1H),8.47(s,1H),7.44(td,J=9.2,8.7,6.4Hz,1H),6.97(ddt,J=11.1,8.5,3.0Hz,2H),5.83(dt,J=11.8,2.7Hz,1H),5.39(ddt,J=14.5,12.0,2.9Hz,2H),4.96(d,J=14.4Hz,1H),4.70–4.57(m,4H),4.52(d,J=5.5Hz,1H),4.10(ddq,J=20.0,5.7,2.9Hz,1H),1.36(d,J=7.3Hz,3H)。
Example 45: preparation of (1S, 2S, 5S) -N- (2, 4-difluorobenzyl) -2- (fluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-A Amide (C45)
(1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (fluoromethyl) -5-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (10 mg,0.19 mmol) was dissolved in 3ml ethanol and 3ml ethyl acetate and sparged under an argon atmosphere. Palladium on carbon (10 wt%, wet) E101 NE/W (2 mg) was added and the mixture was sparged under a hydrogen atmosphere (1 atm, balloon). The mixture was vigorously stirred for one hour, then atAnd spraying under argon atmosphere. By passing throughAnd (5) filtering the pad. Washing ∈10 with absolute ethanol>And the filtrate was concentrated to dryness. The residue was purified by RP-HPLC to give the title product. MS (m/z) 451.200[ M+H ] +1 H NMR (400 MHz, acetonitrile-d 3) delta 10.31 (s, 1H), 8.41 (s, 1H), 7.44 (td, j=9.2, 8.7,6.3hz, 1H), 7.03-6.91 (m, 2H), 5.05-4.26 (m, 7H), 3.62-3.51 (m, 1H), 2.13-2.00 (m, 1H), 1.84 (ddd, j=15.6, 8.0,4.1hz, 1H), 1.77-1.57 (m, 2H), 1.26 (d, j=6.8 hz, 3H).
Example 46: preparation of (1R, 2R, 5S) -8-hydroxy-2-methoxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trio Fluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C46)
Step 1: synthesis of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) Radical) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid
To a suspension of methyl 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylate (10.0 g,17.8mmol,1 eq.) prepared according to example 24 step 3 in methanol (200 mL) and water (100 mL) was added lithium hydroxide monohydrate (5.979 g,142mmol,8 eq.). The reaction mixture was heated to 50 ℃ for 18 hours, diluted with water, and acidified with 1N HCl (aqueous solution). The slurry (2×) was extracted with EtOAc and the combined organic phases were washed with brine, over Na 2 SO 4 Dried, filtered and concentrated to give 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2- Formic acid. MS (m/z) 547.82[ M+H ]] +
Step 2: synthesis of (S) -3- (benzyloxy) -2- (but-3-en-2-ylcarbamoyl) -4-oxo-5- ((2, 4, 6-) Trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) carbamic acid tert-butyl ester
To 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid (6.00 g,11.0mmol,1 eq), (S) -but-3-en-2-amine hydrochloride (1.769 g,16.4mmol,1.5 eq) and HOBt (2.221 g,16.4mmol,1.5 eq) in CH at 0deg.C 2 Cl 2 To a solution of (120 mL) was added DIPEA (9.54 mL,54.8mmol,5 eq.) and EDCI (3.151 g,16.4mmol,1.5 eq.). The reaction mixture was warmed to room temperature and stirred for 18 hours. The reaction mixture was quenched with water and 1M HCl and with CH 2 Cl 2 Extraction (3×). The combined organic phases were taken over N a2 SO 4 Dried, filtered and concentrated. The crude residue was purified by column chromatography (0% -100% etoac/hexanes) and concentrated to give tert-butyl (S) - (3- (benzyloxy) -2- (but-3-en-2-ylcarbamoyl) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) carbamate. MS (m/z) 600.90[ M+H ]] +
Step 3: synthesis of (S) -5- (benzyloxy) -3- (but-3-en-2-yl) -4, 6-dioxo-7- ((2, 4, 6-trifluorobenzyl) Radical) carbamoyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-1-carboxylic acid tert-butyl ester
To a suspension of tert-butyl (S) - (3- (benzyloxy) -2- (but-3-en-2-ylcarbamoyl) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) carbamate (2.20 g,3.66mmol,1 eq.) and cesium carbonate (4.77 g,14.7mmol,4 eq.) in MeCN (55 mL) was added diiodomethane (0.59 mL,7.33mmol,2 eq.). The reaction mixture was heated to 70 ℃ for 8 hours and held with NH 4 Cl (aqueous) quench. The mixture was extracted with EtOAc (2×), and the combined organic phases were taken up over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by column chromatography (0% -100% EtOAc/hexanes) to give (S) -5- (benzyloxy) -3-(but-3-en-2-yl) -4, 6-dioxo-7- ((2, 4, 6-trifluorobenzyl) carbamoyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-1-carboxylic acid tert-butyl ester. MS (m/z) 612.79[ M+H ]] +
Step 4: synthesis of (S) -5- (benzyloxy) -3- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
To (S) -5- (benzyloxy) -3- (but-3-en-2-yl) -4, 6-dioxo-7- ((2, 4, 6-trifluorobenzyl) carbamoyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ][1,2,4]To a solution of t-butyl triazine-1-carboxylate (2.00 g,3.26mmol,1 eq.) in methanol (55 mL) was added aqueous 2N NaOH (2.45 mL,4.90mmol,1.5 eq.). The reaction mixture was heated to 40 ℃ for 1 hour, quenched with 10% citric acid solution, and quenched with CH 2 Cl 2 And (5) diluting. The phases were separated and the aqueous phase was treated with CH 2 Cl 2 And (5) extracting. The combined organic phases were taken up in Na 2 SO 4 Dried, filtered and concentrated. The residue was suspended in 1:1 EtOAc/hexane and the precipitate was collected by filtration to give (S) -5- (benzyloxy) -3- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2, 1-f)][1,2,4]Triazine-7-carboxamide. MS (m/z) 512.96[ M+H ]] +
Step 5: synthesis of 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -1- (1-methoxyallyl) -4, 6-dioxo substituted-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
To (S) -5- (benzyloxy) -3- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]To a solution of triazine-7-carboxamide (0.122 g,0.237mmol,1 eq.) in acetonitrile (5 mL) was added Pd (OAc) 2 (0.0027 g,0.012mmol,0.05 eq.) 1, 3-bis (diphenylphosphine) propane (dppp) (0.0049 g,0.012mmol,0.05 eq.), triethylamine (0.050 mL,0.356mmol,1.5 eq.) and methoxypropadiene (0.100 mL,1.19mmol,5 eq.). The reaction mixture was heated to 100 ℃ for 20 minutes. Water and brine were added and the aqueous phase was extracted with EtOAc (2×). Will be combined with The organic phase is treated by Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by column chromatography (0% -100% etoac/hexanes) to give 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -1- (1-methoxyallyl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide. MS (m/z) 583.00[ M+H ]] +
Step 6: synthesis of (1R, 2R, 5S) -8- (benzyloxy) -2-methoxy-5-methyl-7, 9-dioxo-N- (2, 4,6- Trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxylic acid Amines
To a solution of 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -1- (1-methoxyallyl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (0.112 g,0.192mmol,1 eq.) in 1, 2-dichloroethane (5 mL) was added Hoveyda-Grubbs II catalyst (0.024 g,0.038mmol,0.2 eq.). The reaction mixture was heated to 70 ℃ for 24 hours and concentrated. The residue was purified by preparative HPLC (column, gemini 10. Mu.C 18 110A, AXI/; 250X 21.2 mm) eluting with 5% -100% acetonitrile (0.1% TFA) in water (0.1% TFA) over 20 minutes. The combined fractions were lyophilized to give (1 r,2r,5 s) -8- (benzyloxy) -2-methoxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide. MS (m/z) 554.98.
Step 7: synthesis of (1R, 2R, 5S) -8-hydroxy-2-methoxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluoro) Benzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C46)
To (1R, 2R, 5S) -8- (benzyloxy) -2-methoxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]To a solution of triazacyclononane-10-carboxamide (0.0455 g,0.0821mmol,1 eq.) in DMF (2 mL) was added lithium chloride (0.0348 g, 0.823mmol, 10 eq.). The reaction mixture was heated to 100 ℃ for 1 hour and filtered. Will beThe filtrate was purified by preparative HPLC (column, gemini 10. Mu.C 18 110A, AXI/; 250X 21.2 mm) eluting with 5% -100% acetonitrile (0.1% TFA) in water (0.1% TFA) over 20 minutes. The combined fractions were lyophilized to give (1 r,2r,5 s) -8-hydroxy-2-methoxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 465.02[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δ10.31(t,J=5.8Hz,1H),8.21(s,1H),7.20(t,J=8.6Hz,2H),5.76(dt,J=12.1,2.3Hz,1H),5.55(dt,J=12.0,2.4Hz,1H),5.29–5.17(m,2H),5.06(d,J=14.6Hz,1H),4.75(d,J=14.6Hz,1H),4.57(d,J=5.8Hz,2H),3.39(s,3H),1.28(d,J=7.2Hz,3H)。 19 F NMR(376MHz,DMSO-d 6 )δ-109.25(ddd,J=15.5,9.3,6.2Hz),-112.45–-112.69(m)。
Example 47: preparation of (1R, 2R, 5S) -8-hydroxy-2-methoxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trio Fluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10-carboxylic acid Amine (C47)
Step 1: synthesis of (1R, 2R, 5S) -8-hydroxy-2-methoxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluoro) Benzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxylic acid Amines as well as their use in the treatment of cancer
To (1R, 2R, 5S) -8- (benzyloxy) -2-methoxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 46][1,2,5]To a solution of triazacyclononane-10-carboxamide (0.008 g,0.0144mmol,1 eq.) in methanol (1 mL) was added platinum (IV) oxide (0.0003 g,0.0014 eq. 0.1 eq.). The reaction mixture was evacuated and backfilled with hydrogen (2×), sparged with hydrogen for 5 minutes, and stirred under a hydrogen balloon atmosphere for 1 hour. The reaction mixture was filtered, concentrated and passed through the preparative HPLC purification (column, gemini 10. Mu.C 18110A, AXI/; 250X 21.2 mm) eluting with 5% -100% acetonitrile (0.1% TFA) in water (0.1% TFA) over 20 minutes. The combined fractions were lyophilized to give (1 r,2r,5 s) -8-hydroxy-2-methoxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 467.07[ M+H ] ] +1 H NMR(400MHz,DMSO-d 6 )δ10.37(t,J=5.9Hz,1H),8.25(s,1H),7.20(t,J=8.6Hz,2H),4.69–4.59(m,2H),4.59–4.49(m,2H),4.45(s,1H),4.44–4.35(m,1H),3.53(s,3H),1.91–1.72(m,3H),1.34–1.22(m,1H),1.17(d,J=6.7Hz,3H)。 19 F NMR(376MHz,DMSO-d 6 )δ-109.32(ddd,J=15.5,9.4,6.4Hz),-112.55(t,J=7.2Hz)。
Example 48: preparation of (1S, 5S) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C48)
Step 1: synthesis of (S) -5- (benzyloxy) -3- (but-3-en-2-yl) -1- (2-methylbut-3-en-2-yl) -4,6- dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
To (S) -5- (benzyloxy) -3- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] prepared according to example 46][1,2,4]Triazine-7-carboxamide (0.150 g,0.293mmol,1 eq), (S) -5- (benzyloxy) -3- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide and Pd (PPh) 3 ) 4 To a solution of (0.0338 g,0.029mmol,0.1 eq.) in THF (3 mL) and DMF (0.15 mL) was added methyl (2-methylbut-3-en-2-yl) carbonate (0.0633 g,0.439mmol,1.5 eq.). The reaction mixture was heated to 60 ℃ for 2 hours and concentrated. The residue was purified by column chromatography(0% -100% EtOAc/hexanes) purification and collection of the pure fractions and concentration gave (S) -5- (benzyloxy) -3- (but-3-en-2-yl) -1- (2-methylbut-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ][1,2,4]Triazine-7-carboxamide. MS (m/z) 581.02[ M+H ]] +
Step 2: synthesis of (1S, 5S) -8- (benzyloxy) -2, 5-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide
To a solution of (0.019 g,0.033mmol,1 eq.) in 1, 2-dichloroethane (1 mL) was added Hoveyda-Grubbs II catalyst (0.04 g,0.006mmol,0.2 eq.). The reaction mixture was sprayed with Ar (g) for 10 minutes and heated to 75℃for 18 hours. The mixture was concentrated and purified by column chromatography (0% -100% etoac/hexanes) to give (1 s,5 s) -8- (benzyloxy) -2, 5-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 553.00[ M+H ]] +
Step 3: synthesis of (1S, 5S) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) scheme 2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C49)
(1S, 5S) -8- (benzyloxy) -2, 5-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (0.01 g,0.020mmol,1 eq.) was dissolved in 1:1 TFA/toluene (2 mL) and stirred at room temperature for 5 hours. The reaction mixture was concentrated, dissolved in MeCN, filtered, and purified by preparative HPLC (column, gemini 10 μc18 110a, axi/;250×21.2 mm), eluting with 5% -100% acetonitrile (0.1% tfa) in water (0.1% tfa) over 20 minutes. The combined fractions were lyophilized to give (1S, 5S) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] ][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 463.10[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δ10.33(t,J=5.7Hz,1H),8.25(s,1H),7.23–7.17(m,2H),5.41(td,J=11.8,11.3,2.9Hz,1H),5.33(dd,J=12.4,2.0Hz,1H),5.29–5.18(m,1H),5.06–4.98(m,1H),4.72(d,J=14.5Hz,1H),4.56(d,J=5.6Hz,2H),1.49(s,3H),1.28(d,J=7.2Hz,3H),0.93(s,3H)。 19 F NMR(376MHz,DMSO-d 6 )δ-109.19(ddd,J=15.6,9.3,6.3Hz),-112.52(q,J=7.3,5.9Hz)。
Example 49: preparation of (1S, 10R, 13R) -N- [ (2, 4-difluorophenyl) methyl]-10- (fluoromethyl) -6-hydroxy-) 13-methyl-5, 8-dioxo-1, 2, 9-triazatricyclo [7.4.1.02,7 ]]Tetradec-3,6,11-triene-4-carboxamide (C49 a) and (1S, 10R, 13R) -N- [ (2, 4-difluorophenyl) methyl]-10- (fluoromethyl) -6-hydroxy-13-methyl-5, 8-di Oxo-1, 2, 9-triazatricyclo [7.4.1.02,7 ]]Tetradec-3, 6-diene-4-carboxamide (C49 b)
/>
Step 1: synthesis of 3-benzyloxy-1- [ tert-butoxycarbonyl (1-methallyl) amino group]-5- [ (2, 4-difluorobenzene) Radical) methylcarbamoyl radical]-4-oxo-pyridine-2-carboxylic acid methyl ester
To a mixture of 3-benzyloxy-1- (tert-butoxycarbonylamino) -5- [ (2, 4-difluorophenyl) methylcarbamoyl ] -4-oxo-pyridine-2-carboxylic acid methyl ester (8 g,14.7 mmol) in THF (75 mL) was added but-3-en-2-ol (1.59 g,22.1 mmol), triphenylphosphine (5.79 g,22.1 mmol) at 0 ℃. Diisopropyl azodicarboxylate (4.46 g,22.1 mmol) was then added dropwise. The resulting mixture was stirred at 0 ℃ for 5 minutes, after which it was taken out of the cooling bath and stirred at room temperature for 2 hours. The reaction was mixed with silica gel, concentrated and purified by normal phase chromatography. LCMS-esi+ (m/z): h+ calculated theory for C31H33F2N3O 7: 597.23, found: 597.88.
Step 2: synthesis of 3-benzyloxy-1- [ tert-butoxycarbonyl (1-methallyl) amino group]-5- [ (2, 4-difluorobenzene) Radical) methylcarbamoyl radical]-4-oxo-pyridine-2-carboxylic acid
3-benzyloxy-1- [ tert-butoxycarbonyl (1-methallyl) amino ] -5- [ (2, 4-difluorophenyl) methylcarbamoyl ] -4-oxo-pyridine-2-carboxylic acid methyl ester (10 g,16.7 mmol) was dissolved in a mixture of MeOH (96 mL), THF (48 mL) and water (48 mL). Lithium hydroxide monohydrate (4.2 g,41.96 mmol) was added. The resulting mixture was heated to 60 ℃ for 6 hours. The reaction was then cooled to room temperature, concentrated, the residue diluted with EtOAc, acidified to pH-4 with 1N HCl, the organic layer washed with brine, dried over sodium sulfate, filtered and concentrated to give a pale pink solid. The resulting product was used directly in the next step. LCMS-esi+ (m/z): h+ calculated theory for C30H31F2N3O 7: 583.21, found: 583.87.
step 3: synthesis of N- [ 3-benzyloxy-5- [ (2, 4-difluorophenyl) methylcarbamoyl]-2- [ [ (1R) -1- (hydroxy) Methyl) allyl group]Carbamoyl radicals]-4-oxo-1-pyridinyl]-N- (1-methylallyl) carbamic acid tert-butyl ester
The residue from the previous step (7 g,12.0 mmol) was dissolved in DCM (60 mL) at room temperature. To the stirred mixture was added (2R) -2-aminobut-3-en-1-ol (1.57 g,18.0 mmol), followed by EDCI.HCl (4.12 g,21.6 mmol), HOAt (2.94 g,21.6 mmol) and DIEA (6.2 g,48 mmol). The newly formed mixture was stirred for one hour. The reaction was then diluted with DCM, washed with 10% citric acid, brine, dried over sodium sulfate, filtered and concentrated and used directly in the next step. LCMS-esi+ (m/z): h+ calculated theory for C34H38F2N4O 7: 652.27, found: 653.03.
Step 4: synthesis of 3-benzyloxy-N5- [ (2, 4-difluorophenyl) methyl]-N2- [ (1R) -1- (hydroxymethyl) allyl Base group]-1- (1-methylallylamino) -4-oxo-pyridine-2, 5-dicarboxamide
N- [ 3-benzyloxy-5- [ (2, 4-difluorophenyl) methylcarbamoyl ] -2- [ [ (1R) -1- (hydroxymethyl) allyl ] carbamoyl ] -4-oxo-1-pyridinyl ] -N- (1-methallyl) carbamic acid tert-butyl ester (7.8 g,12 mmol) was dissolved in DCM (70 mL) at room temperature and treated with a 1, 4-dioxane solution of 4N HCl (70 mL) for 1 hour at room temperature. The reaction was concentrated, diluted with EtOAc, slowly basified with saturated sodium bicarbonate until no foaming occurred, then solid sodium bicarbonate was added to saturate the aqueous layer. Extraction with EtOAc, washing of the organic layer with brine, drying over sodium sulfate, filtration and concentration gave a brown oil. The resulting product was used directly in the next step. LCMS-esi+ (m/z): h+ calculated theory for C29H30F2N4O 5: 552.22, found: 553.09.
step 5: synthesis of 5-benzyloxy-N- [ (2, 4-difluorophenyl) methyl]-3- [ (1R) -1- (hydroxymethyl) allyl]- 1- (1-methylallyl) -4, 6-dioxo-2H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
3-benzyloxy-N5- [ (2, 4-difluorophenyl) methyl ] -N2- [ (1R) -1- (hydroxymethyl) allyl ] -1- (1-methallylamino) -4-oxo-pyridine-2, 5-dicarboxamide (1.57 g,2.84 mmol) was partitioned at room temperature into a mixture of DCE (5.3 mL) and acetonitrile (5.3 mL), paraformaldehyde (224 mg) was added. The resulting mixture was then heated to 88 ℃, and a mixture of AcOH (0.79 mL) and TFA (0.79 mL) was added in one portion. The reaction was sealed and stirred for 1 hour. The reaction was then cooled to room temperature, concentrated, redissolved in EtOAc, washed with saturated sodium bicarbonate, brine, dried over sodium sulfate, filtered, and concentrated to a brown oil. The residue was then dissolved in DMF (5 mL) and treated with potassium carbonate (390 mg,2.84 mmol) and benzyl bromide (480 mg,2.84 mmol). The reaction was then heated at 70 ℃ for 2 hours, after which it was cooled to room temperature, partitioned between EtOAc and water. The organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated and purified by normal phase chromatography. LCMS-esi+ (m/z): h+ calculated theory for C30H30F2N4O 5: 564.22, found: 565.02.
Step 6: synthesis of 5-benzyloxy-N- [ (2, 4-difluorophenyl) methyl]-3- [ (1R) -1- (fluoromethyl) allyl]- 1- (1-methylallyl) -4, 6-dioxo-2H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
5-benzyloxy-N- [ (2, 4-difluorophenyl) methyl ] -3- [ (1R) -1- (hydroxymethyl) allyl ] -1- (1-methallyl) -4, 6-dioxo-2H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (851 mg,1.51 mmol) was dissolved in DCM (12.0 mL) at 0deg.C. Bis (2-methoxyethyl) aminothiotrifluoride (1.33 g,6.03 mmol) was added. After addition the reaction was removed from the cooling bath and stirred overnight under ambient conditions. The reaction was then cooled to 0 ℃ and quenched with saturated sodium bicarbonate. Additional sodium bicarbonate powder was added to saturate the mixture. The reaction was extracted with DCM, washed with brine, dried over sodium sulfate, filtered and concentrated and purified by normal phase chromatography. LCMS-esi+ (m/z): h+ calculated theory for C30H29F3N4O 4: 566.21, found: 566.99.
step 7: synthesis of (1S, 10R, 13R) -6-benzyloxy-N- [ (2, 4-difluorophenyl) methyl]10- (fluoromethyl) scheme 13-methyl-5, 8-dioxo-1, 2, 9-triazatricyclo [7.4.1.02,7 ]]Tetradec-3,6,11-triene-4-carboxamide
Spraying 5-benzyloxy-N- [ (2, 4-difluorophenyl) methyl with argon at room temperature ]-3- [ (1R) -1- (fluoromethyl) allyl]-1- (1-methylallyl) -4, 6-dioxo-2H-pyrido [2,1-f][1,2,4]A solution of triazine-7-carboxamide (370 mg,0.65 mmol) in DCE (9 mL). Hoveyda-Grubbs II catalyst HG-M720 (41 mg,0.065 mmol) was added. The mixture was sparged with argon for an additional 5 minutes, then sealed and heated overnight at 80 ℃ under a nitrogen balloon. The reaction was then cooled to room temperature, concentrated and purified by normal phase chromatography. 1 H NMR (400 mhz, cdcl 3) delta 10.38 (t, j=6.0 hz, 1H), 8.54 (s, 1H), 7.55-7.47 (m, 2H), 7.44-7.29 (m, 4H), 6.89-6.76 (m, 2H), 5.83 (dt, j=10.8, 2.3hz, 1H), 5.62-5.45 (m, 3H), 5.25 (d, j=10.5 hz, 1H), 5.11 (d, j=14.6 hz, 1H), 4.79-4.65 (m, 1H), 4.65-4.62 (m, 2H), 4.62-4.46 (m, 1H), 4.26 (d, j=14.6 hz, 1H), 3.77 (tp, j=6.6, 3.3hz, 1H), 1.41 (d, j=6.6 hz, 3H). LCMS-esi+ (m/z): h+ calculated theory for C28H25F3N4O 4: 538.18, found: 539.09.
step 8: synthesis of (1S, 10R, 13R) -N- [ (2, 4-difluorophenyl) methyl]-10- (fluoromethyl) -6-hydroxy-13- Methyl-5, 8-dioxo-1, 2, 9-triazatricyclo [7.4.1.02,7 ]]Tetradec-3,6,11-triene-4-carboxamide
(1S, 10R, 13R) -6-benzyloxy-N- [ (2, 4-difluorophenyl) methyl]-10- (fluoromethyl) -13-methyl-5, 8-dioxo -1,2, 9-triazatricyclo [7.4.1.02,7 ]]Tetradecane-3,6,11-triene-4-carboxamide (20 mg,0.037 mmol) was treated with a mixture of DCM (1.5 mL) and TFA (1.5 mL) at room temperature for 2 hours. The reaction was concentrated, redissolved in DMF and purified by reverse phase preparative HPLC. The absolute configuration of C13 was not confirmed. 1 H NMR (400 MHz, acetonitrile-d 3) delta 10.15 (s, 1H), 8.39 (s, 1H), 7.44 (td, j=9.2, 8.8,6.5hz, 1H), 7.09-6.88 (m, 2H), 5.81 (dt, j=11.5, 2.3hz, 1H), 5.66-5.59 (m, 1H), 5.54-5.41 (m, 1H), 5.13 (d, j=14.5 hz, 1H), 4.80-4.55 (m, 5H), 3.87 (dp, j=10.1, 3.4hz, 1H), 1.37 (d, j=6.7 hz, 3H). LCMS-esi+ (m/z): h+ calculated theory for C21H19F3N4O 4: 448.14, found: 449.10.
step 9: synthesis of (1S, 10R, 13R) -N- [ (2, 4-difluorophenyl) methyl]-10- (fluoromethyl) -6-hydroxy-13- Methyl-5, 8-dioxo-1, 2, 9-triazatricyclo [7.4.1.02,7 ]]Tetradec-3, 6-diene-4-carboxamide
(1S, 10R, 13R) -6-benzyloxy-N- [ (2, 4-difluorophenyl) methyl at room temperature]-10- (fluoromethyl) -13-methyl-5, 8-dioxo-1, 2, 9-triazatricyclo [7.4.1.02,7 ]]Tetradec-3,6,11-triene-4-carboxamide (20 mg,0.037 mmol) was dissolved in EtOH (20 mL). 10% Pd/C (4 mg) was added, the reaction was degassed and flushed with nitrogen three times, then degassed and flushed with hydrogen three times, and then hydrogenated under a hydrogen balloon for 1 hour. The reaction was then degassed and flushed with nitrogen, filtered through celite, concentrated and purified by reverse phase prep HPLC. The absolute configuration of C13 was not confirmed. 1 H NMR (400 MHz, acetonitrile-d 3) delta 10.33 (s, 1H), 8.45 (s, 1H), 7.44 (td, j=9.2, 8.8,6.5hz, 1H), 7.06-6.86 (m, 2H), 4.79-4.39 (m, 7H), 3.63-3.58 (m, 1H), 1.95-1.82 (m, 2H), 1.62 (dt, j=7.1, 3.5hz, 2H), 1.32 (d, j=7.2 hz, 3H). LCMS-esi+ (m/z): h+ calculated theory for C21H21F3N4O 4: 450.15, found: 451.12.
example 50: preparation of (1S, 2R, 5R) -5- (fluoromethyl) -8-hydroxy-2-methyl-7, 9-dioxo-N- (2, 4,6- Trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C50)
Synthesis of 3-benzyloxy-1- [ tert-butoxycarbonyl (1-methallyl) amino group]-4-oxo-5- [ (2, 4, 6-trifluoro) Phenyl) methylcarbamoyl]Pyridine-2-carboxylic acid methyl ester
3-benzyloxy-1- (tert-butoxycarbonylamino) -4-oxo-5- [ (2, 4, 6-trifluorophenyl) methylcarbamoyl at 0℃in THF (200 mL)]To methyl pyridine-2-carboxylate (10 g,17.8 mmol) was added but-3-en-2-ol (1.93 g,26.7 mmol), followed by Ph 3 P (7.0 g,26.7 mmol). DIAD (5.4 g,26.7 mmol) was then added dropwise over 5 minutes. The resulting reaction mixture was stirred at 0 ℃ for 5 minutes and then warmed to room temperature. The reaction was stirred at room temperature overnight. The solvent was removed under vacuum and the resulting crude material was purified by column over silica gel to give the title compound. MS (m/z) 615.99[ M+H ] ] +
Synthesis of 3-benzyloxy-1- [ tert-butoxycarbonyl (1-methallyl) amino group]-4-oxo-5- [ (2, 4, 6-trifluoro) Phenyl) methylcarbamoyl]Pyridine-2-carboxylic acid
To 3-benzyloxy-1- [ tert-butoxycarbonyl (1-methallyl) amino in MeOH (80 mL)]-4-oxo-5- [ (2, 4, 6-trifluorophenyl) methylcarbamoyl]To methyl pyridine-2-carboxylate (11 g,17.9 mmol) was added aqueous LiOH (2.5N) (43 ml,107 mmol). The reaction mixture was heated to 70 ℃ overnight. The reaction mixture was carefully concentrated to remove MeOH. The residue was diluted and rinsed with some water and acidified with 1N HCl to ph=3. EtOAc was added for extraction. The organic phase was separated. The aqueous layer was extracted with more EtOAc. The combined organic phases were washed with water and brine, and dried over Na 2 SO 4 Drying, filtration and concentration gave the title product. The crude material was taken for the next step. MS (m/z) 601.84[ M+H ]] +
Synthesis of N3-Benzyloxy-2- [ [ (1R) -1- (hydroxymethyl) allyl]Carbamoyl radicals]-4-oxo-5- [ (2, 4), 6-trifluorophenyl) methylcarbamoyl]-1-pyridinyl]-N- (1-methylallyl) carbamic acid tert-butyl ester
To 3-benzyloxy-1- [ tert-butoxycarbonyl (1-methallyl) amino group in DMF (50 mL)]-4-oxo-5- [ (2, 4, 6-trifluorophenyl) methylcarbamoyl ]To pyridine-2-carboxylic acid (11.2 g,18.6 mmol) was added DIEA (16.2 ml,93.1 mmol) followed by HATU (9.2 g,24.2 mmol). After the reaction mixture was stirred at room temperature for 1 hour, (2R) -2-aminobut-3-en-1-ol hydrochloride (2.9 g,24.2 mmol) was added to the reaction mixture. After 2 hours, the reaction mixture was diluted with EtOAc and washed with aqueous LiCl. The aqueous layer was extracted with EtOAc (1×). The combined organic phases were washed with water (1×). The organic phase was treated with MgSO 4 Drying and concentration gave the crude product, which was purified by silica gel chromatography to give the title compound. MS (m/z) 670.86[ M+H ]] +
Synthesis of 3-benzyloxy-N2- [ (1R) -1- (hydroxymethyl) allyl]-1- (1-methylallylamino) -4-oxo-) N5- [ (2, 4, 6-trifluorophenyl) methyl ]]Pyridine-2, 5-dicarboxylic acid diamides
N- [ 3-benzyloxy-2- [ [ (1R) -1- (hydroxymethyl) allyl ] at room temperature]Carbamoyl radicals]-4-oxo-5- [ (2, 4, 6-trifluorophenyl) methylcarbamoyl]-1-pyridinyl]Tert-butyl N- (1-methallyl) carbamate (XX-3) (10 g,14.9 mmol) was dissolved in DCM (50 ml). HCl (4M dioxane solution) (11.2 mL,44.7 mmol) was added. The reaction mixture was stirred at room temperature for 24 hours. LCMS showed the reaction was complete. The reaction mixture was then concentrated to dryness. The residue was then partitioned between EtOAc and NaHCO 3 Between the aqueous solutions. The aqueous layer was extracted with EtOAc. The combined organic phases were washed with water, dried over Na2SO4, filtered and concentrated to give the title compound. MS (m/z) 571.06[ M+H ]] +
To 5-hydroxy-3- [ (1R) -1- (hydroxymethyl) allyl]-1- (1-methylallyl) -4, 6-dioxo-N- [ (2), 4, 6-Trifluorophenyl) methyl group]-2H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide (2 g,4.06 mmol) in DMF Adding bromomethylbenzene into the solution
3-benzyloxy-N2- [ (1R) -1- (hydroxymethyl) allyl in CAN (5 ml) and DCE (5 ml) in a 20ml microwave reaction vial]-1- (1-methylallylamino) -4-oxo-N5- [ (2, 4, 6-trifluorophenyl) methyl ]]To pyridine-2, 5-dicarboxamide (XX-4) (0.89 g,1.56 mmol) was added paraformaldehyde (93.1 mg,3.04 mmol), acOH (0.5 mL), followed by TFA (0.5 mL). After addition the vials were capped and heated to 89 ℃ overnight. LCMS showed complete reaction. The reaction mixture was diluted with EtOAc and washed with saturated aqueous NaHCO 3. The organic phase was dried over MgSO4 and the solvent was removed in vacuo to give the title compound as a crude material, which was taken for the next step. MS (m/z) 535.1[ M+H ]] +
Synthesis of [ (2R) -2- [ 5-benzyloxy-1- (1-methallyl) -4, 6-dioxo-7- [ (2, 4, 6-trifluorophenyl) Methylcarbamoyl group]-2H-pyrido [2,1-f][1,2,4]Triazin-3-yl]But-3-enyl]Acetic acid esters
To 5-hydroxy-3- [ (1R) -1- (hydroxymethyl) allyl in DMF]-1- (1-methylallyl) -4, 6-dioxo-N- [ (2, 4, 6-trifluorophenyl) methyl)]-2H-pyrido [2,1-f][1,2,4]To triazine-7-carboxamide (2 g,4.06 mmol) was added bromomethylbenzene (XX-5) (2.08 g,12.2 mmol) followed by potassium carbonate (2.93 g,21.2 mmol). The reaction mixture was stirred at 60 ℃ overnight. Lcms showed complete reaction. The reaction mixture was diluted with EtOAc and washed with saturated aqueous NaHCO 3. The organic phase was dried over MgSO4 and the solvent was removed in vacuo to give the crude material. MS (m/z) 625[ M+H ]] +
Synthesis of 5-benzyloxy-3- [ (1R) -1- (hydroxymethyl) allyl]1- (1-methylallyl) -4, 6-dioxo-N-) [ (2, 4, 6-Trifluorophenyl) methyl group]-2H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
To crude [ (2R) -2- [ 5-benzyloxy-1- (1-methallyl) -4, 6-dioxo-7- [ (2, 4, 6-trifluorophenyl) methylcarbamoyl ] in MeOH (100 mL)]-2H-pyrido [2,1-f][1,2,4]Triazin-3-yl]But-3-enyl]NaOH (1N) (63 ml,63.3 mmol) was added to the acetate (7.61 g,12.2 mmol). The mixture was stirred at room temperature for 30 minutes. LCMS showed completion of the reaction. The solvent was removed in vacuo and the resulting residue was diluted in EtOAc and taken up with H 2 And (3) washing. The organic phase was treated with MgSO 4 Dried and concentrated in vacuo. The resulting crude material was purified by a silicone gel column to afford the title compound. MS (m/z) 583.01[ M+H ]] +
Synthesis of 5-benzyloxy-3- [ (1R) -1- (fluoromethyl) allyl]1- (1-methylallyl) -4, 6-dioxo-N-) [ (2, 4, 6-Trifluorophenyl) methyl group]-2H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
To 5-benzyloxy-3- [ (1R) -1- (hydroxymethyl) allyl in DCM (8 ml) at 0deg.C]-1- (1-methylallyl) -4, 6-dioxo-N- [ (2, 4, 6-trifluorophenyl) methyl)]-2H-pyrido [2,1-f][1,2,4]To triazine-7-carboxamide (XX-7) (0.82 g,1.41 mmol) was added deoxo-fluor (2.7N) (5.21 ml,14.1 mmol) in toluene, and the mixture was then warmed slowly to room temperature and stirred overnight. The reaction mixture was slowly added to ice-cold NaHCO 3 In aqueous solution, then extracted with DCM. The DCM phase was treated with MgSO 4 The crude product was dried and purified by passing through a silicone gel column to give the title compound. MS (m/z) 585.03[ M+H ]] +
Synthesis of (1S, 10R, 13R) -6-benzyloxy-10- (fluoromethyl) -13-methyl-5, 8-dioxo-N- [ (2, 4, 6-trio-ne) Fluorophenyl) methyl group]-1,2, 9-triazatricyclo [7.4.1.02,7 ]]Tetradec-3,6,11-triene-4-carboxamide
5-benzyloxy-3- [ (1R) -1- (fluoromethyl) allyl ]-1- (1-methylallyl) -4, 6-dioxo-N- [ (2, 4, 6-trifluorophenyl) methyl)]-2H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide (XX-9) (0.41 g,0.7 mmol) was dissolved in DEC (25 ml) and degassed by vacuum, then refilled with N2 (. Times.3) at room temperature. Catalyst HG-II was then added to the solution and flushed with argon with a vent needle at 80℃for 1.5 hours. Then, the vent needle was removed and the reaction was stirred overnight at 80 ℃ with an argon balloon. LCMS showed complete reaction, solvent was removed in vacuo and the resulting residue was purified by a silicone gel column to give the title compound (XX-9). MS (m/z) 556.94[ M+H ]] +
Synthesis of the synthesis (1 s,2r,5R) -5- (fluoromethyl) -8-hydroxy-2-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C50)
To (1S, 10R, 13R) -6-benzyloxy-10- (fluoromethyl) -13-methyl-5, 8-dioxo-N- [ (2, 4, 6-trifluorophenyl) methyl ] in toluene (2 mL)]-1,2, 9-triazatricyclo [7.4.1.02,7 ]]To tetradec-3,6,11-triene-4-carboxamide (80 mg,0.144 mmol) was added TFA (2 ml), and the reaction mixture was stirred at room temperature overnight. LCMS showed complete reaction. The solvent was removed in vacuo and the resulting material was purified by prep HPLC to give the title compound (51). MS (m/z) 467[ M+H ] +1 H NMR (400 MHz, chloroform-d) delta 10.19 (s, 1H), 8.57 (s, 1H), 6.75-6.68 (m, 3H), 5.88 (dt, j=11.4, 2.3hz, 1H), 5.65 (dt, j=11.4, 3.5hz, 1H), 5.49 (dd, j=32.6, 3.1hz, 1H), 5.26 (d, j=14.6 hz, 1H), 4.84 (dd, j=9.9, 3.3hz, 1H), 4.77-4.66 (m, 3H), 4.67-4.55 (m, 2H), 3.80 (dq, j=6.7, 3.4hz, 1H), 1.43 (d, j=6.7 hz, 3H).
Example 51: preparation of (1S, 2R, 5R) -5- (fluoromethyl) -8-hydroxy-2-methyl-7, 9-dioxo-N- (2, 4,6- Trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-A Amide (C51)
(1S, 10R, 13R) -6-benzyloxy-10- (fluoromethyl) -13-methyl-5, 8-dioxo-N- [ (2, 4, 6-trifluorophenyl) methyl]-1,2, 9-triazatricyclo [7.4.1.02,7 ]]Tetradec-3,6,11-triene-4-carboxamide (from example 50) (520 mg,0.93 mmol) was dissolved in EtOH (100 ml) followed by Pd/C (10% by weight) (7.8 mg,0.09 mmol). The mixture was then evacuated and treated with H 2 Backfilling for three times. Reaction in H 2 Stir overnight. LCMS showed complete reaction. The reaction mixture was filtered through celite to remove Pd/C and the solvent was removed under vacuum. The crude material was purified by preparative HPLC to give the title compound (XX). MS (MS)(m/z)469.11[M+H] + 1H NMR (400 MHz, chloroform-d) δ10.37 (s, 1H), 8.61 (s, 1H), 6.82-6.59 (m, 2H), 4.79-4.44 (m, 7H), 3.64-3.39 (m, 1H), 2.35-2.27 (m, 1H), 2.14-1.86 (m, 1H), 1.71-1.61 (m, 2H), 1.38 (d, J=7.1 Hz, 3H).
Example 52: preparation of (1S, 2R,4S, 5S) -N- (2, 4-difluorobenzyl) -4-fluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-A Amide (C52)
Step 1: preparation of (1S, 2R,4R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-hydroxy-2, 5-dimethyl 1, 2-b-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2 ]][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 29][1,2,5]A solution of triazacyclononane-10-carboxamide (45 mg,0.85 mmol) in isopropyl alcohol (5 mL) was purged with argon. To this solution were added phenylsilane (189 mg,1.75 mmol) and tris (2, 6-tetramethyl-3, 5-heptanedionato) manganese (III) (16 mg,0.026 mmol). The reaction mixture was stirred at room temperature under an oxygen balloon for one day. The reaction was then quenched by addition of 10% sodium thiosulfate solution and the mixture extracted with EtOAc. The organic phase was separated and passed over MgSO 4 Drying, filtration, concentration and purification of the residue by chromatography on silica gel eluting with 0% to 100% hexanes/EtOAc afforded the title product. MS (m/z) 539.03[ M+H ] ]+。
Step 2: preparation of (1S, 2R,4S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-fluoro-2, 5-dimethyl- 7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10- Formamide
To (1S, 2R,4R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] at 0 DEG C][1,2,5]To a solution of triazacyclononane-10-carboxamide (20 mg,0.037 mmol) in DCM (2 mL) was added a solution of deoxyfluor in toluene (50%, 0.041mL,0.11 mmol). The reaction mixture was stirred at 0deg.C for 15 min and purified by addition of saturated NaHCO 3 The reaction was quenched with solution. The mixture was extracted with DCM and the organic phase separated and taken up over MgSO 4 Drying, filtration, concentration and purification by silica gel chromatography (eluting with 0% -100% hexanes/EtOAc) afforded the title compound. MS (m/z) 540.92[ M+H ]]+。
Step 3: preparation of (1S, 2R,4S, 5S) -N- (2, 4-difluorobenzyl) -4-fluoro-8-hydroxy-2, 5-dimethyl-7, 9- Dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxylic acid Amines
To (1S, 2R,4S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-fluoro-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ][1,2,5]To a solution of triazacyclononane-10-carboxamide (13 mg,0.024 mmol) in DCM (1 mL) was added TFA (1 mL). The reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated and the residue was purified by reverse phase HPLC eluting with 5% to 100% aqueous acetonitrile to give the title compound. MS (m/z) 451.12[ M+H ]] + .1H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 7.45 (td, J=8.5, 6.4Hz, 1H), 7.04-6.90 (m, 2H), 5.13-5.03 (m, 1H), 5.03-4.91 (m, 2H), 4.73 (d, J=15.0 Hz, 1H), 4.65 (s, 2H), 3.50-3.41 (m, 1H), 2.42 (dt, J=15.8, 7.7Hz, 1H), 2.01-1.84 (m, 1H), 1.52 (dd, J=7.1, 1.7Hz, 3H), 1.41 (dd, J=7.1, 2.7Hz, 3H).
Example 53: preparation of (1S, 2R,4R, 5S) -N- (2, 4-difluorobenzyl) -4-fluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-A Amide (C53)
Step 1: preparation of (1S, 2R,4S, 5S) -8- (benzyloxy) -10- ((2, 4-difluorobenzyl) carbamoyl) -2,5- Dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclonones Alkan-4-yl benzoate
To (1S, 2R,4R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 52 ][1,2,5]To a solution of triazacyclononane-10-carboxamide (36.0 mg,0.067 mmol) in THF (2 mL) was added benzoic acid (24.5 mg,0.201 mmol), diisopropyl azodicarboxylate (40.6 mg,0.201 mmol) and triphenylphosphine (53 mg,0.201 mmol). The reaction mixture was stirred at room temperature for 3 hours. The reaction mixture was diluted with EtOAc, saturated NaHCO 3 Washed and extracted with EtOAc. The organic phase was dried over MgSO 4 Drying, filtration, concentration and purification of the residue by silica gel chromatography eluting with 0% to 100% hexanes/EtOAc afforded the title product. MS (m/z) 643.02[ M+H ]]+。
Step 2: preparation of (1S, 2R,4S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-hydroxy-2, 5-dimethyl 1, 2-b-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2 ]][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide
To (1S, 2R,4S, 5S) -8- (benzyloxy) -10- ((2, 4-difluorobenzyl) carbamoyl) -2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]To a solution of triazacyclononan-4-yl benzoate (15 mg,0.023 mmol) in MeOH (2 mL) and water (0.5 mL) was added LiOH H 2 O (2.80 mg,0.117 mmol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated. The residue was taken up in saturated NaHCO 3 Wash, extract with EtOAc, over MgSO 4 Dried, filtered, concentrated and used for the next step without purification.
Step 3-4: preparation of (1S, 2R,4R, 5S) -N- (2, 4-difluoro)Benzyl) -4-fluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-A Amides and their use
(1S, 2R,4R, 5S) -N- (2, 4-difluorobenzyl) -4-fluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide prepared in analogy to example 52 except that (1S, 2R,4R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-fluoro-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] is used][1,2,5]Triazacyclononane-10-carboxamide replaces (1S, 2R,4S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-fluoro-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Other than triazacyclononane-10-carboxamide. MS (m/z) 451.11[ M+H ]] + .1H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 7.46 (dd, J=8.7, 6.4Hz, 1H), 7.04-6.90 (m, 2H), 5.13-5.03 (m, 1H), 5.03-4.91 (m, 2H), 4.72 (d, J=15.0 Hz, 1H), 4.65 (s, 2H), 3.46 (d, J=8.4 Hz, 1H), 2.42 (dt, J=15.7, 7.9Hz, 1H), 2.00-1.84 (m, 2H), 1.52 (dd, J=7.0, 1.7Hz, 3H), 1.41 (dd, J=7.0, 2.7Hz, 3H).
Example 54: preparation of (1S, 2R, 5S) -N- (2, 4-difluorobenzyl) -4, 4-difluoro-8-hydroxy-2, 5-dimethyl-) 7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10- Formamide (C54)
Step 1: preparation of (1S, 2R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-4, 7, 9-tris Oxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
To (1S, 2R,4R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2- ] at 0deg.Cb][1,2,5]To a solution of triazacyclononane-10-carboxamide (60 mg,0.11 mmol) in DCM (2.0 mL) was added dess-Martin periodate (94.5 mg,0.223 mmol). The reaction mixture was then stirred at room temperature for 3 hours. By adding 1N NaS 2 SO 3 The reaction mixture was quenched with saturated NaHCO 3 Washed and extracted with DCM. The organic phase was separated over MgSO 4 Drying, filtration, concentration and purification of the residue by silica gel chromatography eluting with 0% to 100% hexanes/EtOAc afforded the title compound. MS (m/z) 537.09[ M+H ]]+。
Step 2: preparation of (1S, 2R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 4-difluoro-2, 5-dimethyl-) 7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10- Formamide
To (1S, 2R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-4, 7, 9-trioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] at 0 DEG C][1,2,5]To a solution of triazacyclononane-10-carboxamide (35 mg,0.065 mmol) in DCM (2.0 mL) was added a solution of deoxyfluor in toluene (50%, 0.072mL,0.20 mmol). The reaction was stirred at room temperature overnight. To the reaction mixture was added more deoxyfluor in toluene (50%, 0.072ml,0.20 mmol) and stirred at room temperature. After one day, more deoxyfluor in toluene (50%, 0.072ml,0.20 mmol) was added to the mixture. The reaction mixture was stirred at room temperature for about 2 weeks. By addition of saturated NaHCO 3 The reaction was quenched and extracted with DCM. The organic phase was separated over MgSO 4 Drying, filtration, concentration and purification of the residue by silica gel chromatography eluting with 0% to 100% hexanes/EtOAc afforded the title compound. MS (m/z) 559.09[ M+H ]]+。
Step 3: preparation of (1S, 2R, 5S) -N- (2, 4-difluorobenzyl) -4, 4-difluoro-8-hydroxy-2, 5-dimethyl-7, 9-) Dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] ][1,2,5]Triazacyclononane-10-carboxylic acid Amines
(1S, 2R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 4-difluoro-2,5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]A solution of triazacyclononane-10-carboxamide (13 mg,0.023 mmol) in DCM (1 mL) and TFA (1 mL) was stirred overnight at room temperature. The reaction mixture was concentrated and the residue was purified by reverse phase HPLC eluting with 5% to 100% acetonitrile/water to give the title compound. MS (m/z) 469.26[ M+H] + .1H NMR (400 MHz, chloroform-d) δ10.23 (t, J=5.7Hz, 1H), 8.56 (s, 1H), 7.38 (td, J=8.7, 6.4Hz, 1H), 6.90-6.78 (m, 2H), 5.06 (dp, J=14.1, 7.1Hz, 1H), 4.83-4.56 (m, 4H), 3.37 (tt, J=7.9, 5.1Hz, 1H), 2.42-2.21 (m, 2H), 1.52 (dd, J=7.0, 1.7Hz, 3H), 1.44 (dd, J=7.3, 2.6Hz, 3H).
Example 55: preparation of (1S, 2R, 5S) -4-fluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trio) Fluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C55)
(1S, 2R, 5S) -4-fluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide is prepared in analogy to preparation 54, except that (1S, 2R,4R, 5S) -8- (benzyloxy) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] is used ][1,2,5]Triazacyclononane-10-carboxamide replaces (1S, 2R,4R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide and (1S, 2R, 5S) -8- (benzyloxy) -4-fluoro-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide is a secondary product of the fluorination reaction. MS (m/z) 467.17[ M+H] + .1H NMR (400 MHz, methanol-d 4) δ10.47 (s, 1H), 8.45 (s, 1H), 6.92 (t, J=8.4 Hz, 2H), 5.53 (d, J=7.7 Hz, 1H), 5.33-5.14(m,2H),4.79(d,J=14.6Hz,1H),4.69(t,J=3.5Hz,2H),3.92(dq,J=6.9,3.5Hz,1H),1.49(dd,J=7.3,1.8Hz,3H),1.44(d,J=6.8Hz,3H)。
Example 56: preparation of (1S, 2R,4R, 5S) -4- (difluoromethyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo- N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclic ring Nonane-10-carboxamide (C56)
Step 1: preparation of (1S, 2R,5S, E) -8- (benzyloxy) -4- (methoxymethylene) -2, 5-dimethyl-7, 9-di- oxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Trinitrogen Heterocyclononane-10-carboxamide
To (1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-4, 7, 9-trioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] at 0 DEG C ][1,2,5]To a solution of triazacyclononane-10-carboxamide (120 mg,0.22 mmol) in methanol (1.5 mL) was added 1-diazo-1-dimethoxyphosphoryl-propan-2-one (96 mg,0.5 mmol) and potassium tert-butoxide (85 mg,0.76 mmol). The reaction mixture was stirred at 0 ℃ for 45 minutes. The reaction mixture was diluted with EtOAc, saturated NaHCO 3 Washing, extraction with EtOAc, separation of the organic phase, and removal of the organic phase over MgSO 4 Drying, filtration, concentration and purification by column on silica gel eluting with 0% to 100% hexanes/EtOAc afforded the title compound. MS (m/z) 583.06[ M+H ]]+。
Step 2: preparation of (1S, 2R, 5S) -8- (benzyloxy) -4-formyl-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide
(1S, 2R,5S, E) -8- (benzyloxy) -4- (methoxymethylene) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (30 mg,0.051 mmol) was treated with THF (1.5 mL) and 1N HCl (1.5 mL) in a microwave vial. The vials were then sealed and heated to 55 ℃ for 1 day. The reaction was quenched with saturated sodium bicarbonate solution and extracted with EtOAc. The organic phase was washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography eluting with 0% to 100% etoac/hexanes. MS (m/z) 569.07[ M+H ] +.
Step 3: preparation of (1S, 2R,4R, 5S) -8- (benzyloxy) -4- (difluoromethyl) -2, 5-dimethyl-7, 9-dioxo substituted-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triaza-s Cyclononane-10-carboxamide
To (1S, 2R, 5S) -8- (benzyloxy) -4-formyl-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] at 0deg.C][1,2,5]To a solution of triazacyclononane-10-carboxamide (15 mg,0.026 mmol) in DCM (2.0 mL) was added a solution of deoxyfluor in toluene (50%, 23mg,0.053 mmol). The reaction mixture was stirred at 0 ℃ for 1 hour. By slow addition of saturated NaHCO at 0deg.C 3 The reaction was quenched and extracted into DCM. The organic phase was separated over MgSO 4 Dried, filtered, concentrated and the residue purified by silica gel chromatography (0% -100% etoac/hexanes) to give the title compound. MS (m/z) 591.06[ M+H ]]+。
Step 4: preparation of (1S, 2R,4R, 5S) -4- (difluoromethyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclonones Alkane-10-carboxamides
(1S, 2R,4R, 5S) -8- (benzyloxy) -4- (difluoromethyl) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ][1,2,5]A solution of triazacyclononane-10-carboxamide (5 mg,0.0085 mmol) in DCM (0.5 mL) and TFA (0.5 mL) was stirred overnight at room temperature. The reaction mixture was concentrated and the residue was purified by reverse phase HPLCEluting with 5% -100% acetonitrile/water to give the title compound. MS (m/z) 501.22[ M+H ]] + .1H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 6.98-6.85 (m, 2H), 6.02 (td, J=56.0, 2.8Hz, 1H), 4.83-4.59 (m, 5H), 3.72 (d, J=7.6 Hz, 1H), 2.47 (td, J=8.3, 7.7,2.8Hz, 1H), 1.68 (dd, J=6.1, 3.3Hz, 2H), 1.47 (d, J=6.8 Hz, 3H), 1.42 (d, J=7.2 Hz, 3H).
Example 57: preparation of (1S, 2R,4R, 5S) -4- (fluoromethyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclonones Alkane-10-carboxamide (C57)
Step 1: preparation of (1S, 2R,4R, 5S) -8- (benzyloxy) -4- (hydroxymethyl) -2, 5-dimethyl-7, 9-dioxo- N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclic ring Nonane-10-carboxamide
To (1S, 2R, 5S) -8- (benzyloxy) -4-formyl-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] at 0deg.C ][1,2,5]To a solution of triazacyclononane-10-carboxamide (20 mg,0.035 mmol) in MeOH (5 mL) was slowly added sodium borohydride (0.65 mg,0.017 mmol). The reaction mixture was stirred at 0 ℃ for 15 minutes. After the reaction is completed, saturated NaHCO is added 3 The reaction was quenched and extracted with DCM. The organic phase was separated over MgSO 4 Dried, filtered, concentrated and the residue was used in the next step without purification. MS (m/z) 571.22[ M+H]+。
Step 2-3: preparation of (1S, 2R,4R, 5S) -4- (fluoromethyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclonones Alkane-10-carboxamides
(1S,2R,4R,5S)-4- (fluoromethyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide is prepared in analogy to example 56 except that (1S, 2R,4R, 5S) -8- (benzyloxy) -4- (hydroxymethyl) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b is used][1,2,5]Triazacyclononane-10-carboxamide replaces (1S, 2R, 5S) -8- (benzyloxy) -4-formyl-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Other than triazacyclononane-10-carboxamide. MS (m/z) 483.29[ M+H ]] + .1H NMR (400 MHz, methanol-d 4) delta 8.48 (s, 1H), 6.97-6.85 (m, 2H), 4.87-4.72 (m, 2H), 4.69 (d, J=7.3 Hz, 2H), 4.59-4.30 (m, 3H), 3.71-3.63 (m, 1H), 2.28 (dddd, J=21.6, 16.5,10.9,5.9Hz, 1H), 1.73 (ddd, J=14.5, 10.7,3.4Hz, 1H), 1.61 (dd, J=15.5, 3.2Hz, 1H), 1.42 (dd, J=7.0, 4.4Hz, 6H).
Example 58: preparation of (1S, 2S,4S, 5S) -N- (2, 4-difluorobenzyl) -4-fluoro-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-A Amide (C58)
Step 1: preparation of (1S, 2S,4R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-hydroxy-2, 5-dimethyl 1, 2-b-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2 ]][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide
A solution of (1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (0.576 mmol,300 mg) in i-PrOH (14 mL) from example 30 was sparged with argon for 10 minutes, then treated with phenylsilane (2 eq., 1.15mmol, 140. Mu.L) and tris (2, 6-tetramethyl-3, 5-heptanedione) manganese (III) (0.03 eq., 0.017mmol,10.5 mg) and an oxygen balloon was immobilized. The reaction mixture was stirred at room temperature overnight, at which time additional phenylsilane (1 eq, 0.576mmol,70 μl) and tris (2, 6-tetramethyl-3, 5-heptanedione) manganese (III) (0.03 eq, 0.017mmol,10.5 mg) were added. After stirring for an additional 24 hours, the reaction mixture was quenched with 10% sodium thiosulfate and extracted into EtOAc (2×). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The crude residue was purified by silica gel chromatography (0% -100% etoac in hexanes) to give (1 s,2s,4r,5 s) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide and (1 s,2s,5 s) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-4, 7, 9-trioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide. Alcohol: MS (m/z) 539.15[ M+H ] +. Ketone: MS (m/z) 537.07[ M+H ] +.
Step 2: preparation of (1S, 2S,4S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-fluoro-2, 5-dimethyl- 7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10- Formamide
A solution of (1S, 2S,4R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (0.033 mmol,18 mg) in CH2Cl2 (0.3 mL) was treated with [ bis (2-methoxyethyl) amino ] sulfur trifluoride (2.7M in toluene, 2 eq, 0.067mmol, 25. Mu.L) and sealed. After stirring at room temperature for one hour, the reaction mixture was carefully quenched with saturated sodium bicarbonate and then extracted into EtOAc (3×). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The crude residue was continued without further purification. MS (m/z) 541.16[ M+H ] +.
Step 3: preparation of (1S, 2S,4S, 5S) -N- (2, 4-difluorobenzyl) -4-fluoro-8-hydroxy-2, 5-dimethyl-7, 9- Dioxo-2, 3,4,5,79-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxylic acid Amines
The solution of crude (1S, 2S,4S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4-fluoro-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide was treated with toluene (0.5 mL) and trifluoroacetic acid (0.5 mL) and then stirred at room temperature for 7 hours. The reaction mixture was concentrated and purified by preparative HPLC (10% -100% mecn in water, 0.1% tfa) and then lyophilized to give the title compound. MS (m/z) 451.23[ M+H ] +.1H NMR (400 MHz, methanol-d 4) delta 8.47 (s, 1H), 7.44 (td, J=8.4, 6.3Hz, 1H), 7.01-6.88 (m, 2H), 5.08 (ddd, J=48.4, 6.7,5.0Hz, 1H), 4.97-4.89 (m, 1H), 4.89-4.75 (m, 2H), 4.63 (s, 2H), 4.01 (ddt, J=14.6, 7.8,3.9Hz, 1H), 2.20 (ddd, J=16.1, 6.7,1.8Hz, 1H), 1.67 (ddd, J=35.4, 16.1,11.7Hz, 1H), 1.42 (dd, J=7.2, 2.3Hz, 3H), 1.19 (d, J=7.0 Hz, 3H).
Example 59: preparation of (1S, 2S, 5S) -N- (2, 4-difluorobenzyl) -4, 4-difluoro-8-hydroxy-2, 5-dimethyl-) 7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10- Formamide C (59)
Step 1: preparation of (1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 4-difluoro-2, 5-dimethyl- 7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10- Formamide
(1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-4, 7, 9-trioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 58 in a polypropylene pipe][1,2,5]Triazacyclononane-10-carboxamide (0.065 mmol,35 mg) in CH 2 Cl 2 The solution in (0.65 mL) was treated with [ bis (2-methoxyethyl) amino ]]Sulfur trifluoride (2.7M toluene solution,2 equivalents, 0.13mmol,50 μl) was treated and sealed. After stirring overnight at room temperature, one more part of [ bis (2-methoxyethyl) amino group was added]Sulfur trifluoride (2.7M toluene solution, 2 eq, 0.13mmol, 50. Mu.L). After a further 3 hours, the reaction mixture was carefully quenched with saturated sodium bicarbonate and then extracted into EtOAc (3×). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The crude residue was continued without further purification. MS (m/z) 559.10[ M+H ] ]+。
Step 2: preparation of (1S, 2S, 5S) -N- (2, 4-difluorobenzyl) -4, 4-difluoro-8-hydroxy-2, 5-dimethyl-7, 9-) Dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxylic acid Amines
The solution of crude (1 s,2s,5 s) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 4-difluoro-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide was treated with toluene (1 mL) and trifluoroacetic acid (1 mL) and then stirred at room temperature for 7 hours. The reaction mixture was concentrated and purified by preparative HPLC (10% -100% mecn in water, 0.1% tfa) and then lyophilized to give the title compound. MS (m/z) 469.23[ M+H ] +.1H NMR (400 MHz, methanol-d 4) delta 8.45 (s, 1H), 7.50-7.41 (m, 1H), 7.03-6.91 (m, 2H), 5.08-4.96 (m, 1H), 4.97-4.79 (m, 2H), 4.65 (s, 2H), 4.04-3.92 (m, 1H), 2.41-2.10 (m, 2H), 1.45 (dd, J=7.2, 2.0Hz, 3H), 1.23 (d, J=6.9 Hz, 3H).
Example 60: preparation of (4R, 5S, 13S) -N- (2, 4-difluorobenzyl) -10-hydroxy-4, 13-dimethyl-9, 11-di- oxo-4,9,11,13-tetrahydro-1H-5, 12-methanoimidazo [4,5-g]Pyrido [1,2-b][1,2,5]Triazacyclic ring Nonane-8-carboxamide (C60)
Step 1: preparation of (1S, 2R,3R,4S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -3, 4-dihydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclic ring Nonane-10-carboxamide
A solution of (1S, 2R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (0.48 mmol,250 mg) in acetone (10 mL) and water (1.25 mL) prepared according to example 29 was cooled to 0℃and then treated with N-methylmorpholine N-oxide solution (50% by weight in water, 1.1 eq, 0.528mmol, 110. Mu.L) and osmium tetroxide solution (2.5% by weight in t-butanol, 0.04 eq, 0.019mmol, 195. Mu.L). The reaction mixture was allowed to warm slowly to room temperature and stirred for 4 days, then quenched with 10% aqueous sodium sulfite and extracted into 1/1EtOAc/n-BuOH (2X). The combined organic layers were washed with saturated aqueous sodium bicarbonate and brine, then dried over magnesium sulfate, filtered and concentrated. The crude residue was purified by silica gel chromatography (0-20% meoh in DCM). MS (m/z) 555.12[ M+H ] +.
Step 2: preparation of (1S, 2R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-3, 4,7,9- Tetraoxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] ][1,2,5]Triazacyclononane-10-carboxylic acid Amines
(1S, 2R,3R,4S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -3, 4-dihydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (0.283 mmol,157 mg) in CH 2 Cl 2 The solution in (3 mL) was treated with dess-Martin periodate (3 eq, 0.85mmol,360 mg). The reaction mixture was quenched with 10% sodium thiosulfate, extracted into EtOAc, washed with saturated sodium bicarbonate (3×) and brine, then dried over sodium sulfate, filtered and concentrated. The crude residue was purified by silica gel chromatography (0-20% meoh in DCM). MS (m/z) 569.18[ M+H ] 3 O]+。
Step 3: preparation of (4R, 5S, 13S) -10- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 13-dimethyl-9, 11-) Dioxo-49,11, 13-tetrahydro-1H-5, 12-methanoimidazo [4,5-g]Pyrido [1,2-b][1,2,5]Triaza-s Cyclononane-8-carboxamide
A solution of (1S, 2R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-3, 4,7, 9-tetraoxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (0.054 mmol,30 mg) in EtOH (0.5 mL) was treated with paraformaldehyde (1.5 eq, 0.082mmol,7.4 mg), ammonium acetate (3 eq, 0.16mmol,12.6 mg) and acetic acid (0.5 eq, 0.027mmol, 1.6. Mu.L) and then stirred at room temperature for 72 hours. The reaction was then quenched with saturated aqueous sodium bicarbonate and extracted into EtOAc (3×). The combined organic layers were dried over sodium sulfate, filtered and concentrated. The crude residue was carried forward without further purification. MS (m/z) 561.23[ M+H ] +.
Step 4: preparation of (4R, 5S, 13S) -N- (2, 4-difluorobenzyl) -10-hydroxy-4, 13-dimethyl-9, 11-dioxo substituted-4,9,11,13-tetrahydro-1H-5, 12-methanoimidazo [4,5-g]Pyrido [1,2-b][1,2,5]Triazacyclonones Alkyl-8-carboxamides
Crude (4R, 5S, 13S) -10- (benzyloxy) -N- (2, 4-difluorobenzyl) -4, 13-dimethyl-9, 11-dioxo-4,9,11,13-tetrahydro-1H-5, 12-methanoimidazo [4,5-g]Pyrido [1,2-b][1,2,5]The solution of triazacyclononane-8-carboxamide was treated with toluene (1 mL) and trifluoroacetic acid (1 mL) and then stirred at room temperature overnight. The reaction mixture was concentrated and purified by preparative HPLC (10% -100% mecn in water, 0.1% tfa) and then lyophilized to give the title compound. MS (m/z) 471.21[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d 3) delta 10.12 (t, j=5.3 hz, 1H), 8.47 (s, 1H), 8.32 (s, 1H), 7.47-7.37 (m, 1H), 7.01-6.90 (m, 2H), 5.86 (q, j=7.0 hz, 1H), 5.06 (d, j=14.9 hz, 1H), 4.79 (d, j=14.9 hz, 1H), 4.59 (d, j=6.0 hz, 2H), 4.55-4.45 (m, 1H), 1.65 (d, j=6.8 hz, 3H), 1.60 (d, j=7.2 hz, 3H).
Example 61: preparation of (1S, 2R,3S,4R, 5S) -8-hydroxy-3, 4-dimethoxy-2, 5-dimethyl-7, 9-dioxo substituted-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] ][1,2,5]Triaza-s Cyclononane-10-carboxamide (C61)
(1S, 2R,3S,4R, 5S) -8- (benzyloxy) -3, 4-dimethoxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] prepared in analogy to example 32][1,2,5]Triazacyclononane-10-carboxamide (20 mg,0.033 mmol) was dissolved in MgBr 2 (18.4 mg,0.1 mmol) CH 3 CN (1.2 mL) and stirred at 50deg.C for 2 hours. The reaction was concentrated and purified via preparative HPLC eluting with 10% -100% acetonitrile (0.1% tfa) in water (0.1% tfa) to give the title compound. MS (m/z): 511.1[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d) 3 )δ10.44(s,1H),8.40(s,1H),6.98–6.73(m,2H),4.67–4.56(m,2H),4.50(t,J=14.8Hz,2H),4.21-4.16(m,1H),3.80(d,J=7.1Hz,1H),3.61(d,J=3.1Hz,1H),3.46(d,J=8.8Hz,1H),3.36(s,3H),3.09(s,3H),1.37(t,J=7.6Hz,6H)。
Example 62: preparation of (1S, 2R, 5S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 3, 5-trimethyl-7, 9-dioxo Substituted-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C62)
Step 1: preparation of (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -5- ((2, 4-difluorobenzyl) Yl) carbamoyl) -4-oxopyridin-1 (4H) -yl) (3-methylbut-3-en-2-yl) carbamic acid tert-butyl ester
Tert-butyl (S) - (3- (benzyloxy) -2- (but-3-en-2-ylcarbamoyl) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxopyridin-1 (4H) -yl) carbamate (700 mg,1.20 mmol) prepared according to example 29 was dissolved in THF (6.0 mL). 3-Methylbut-3-en-2-ol (186 mg,2.16 mmol) and triphenylphosphine (553mg, 2.10 mmol) were added. Diisopropyl azodicarboxylate (0.423 mL,2.04 mmol) was added over a period of minutes at ambient temperature. After 1 hour, the reaction was concentrated by rotary evaporation and the residue was purified by flash chromatography (hexane: etOAc) to give tert-butyl (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxopyridin-1 (4H) -yl) (3-methylbutan-3-en-2-yl) carbamate. ES/MS 651.023 (M+H+).
Step 2: preparation of 3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -N5- (2, 4-difluorobenzyl) -1- ((3-methyl) Monobut-3-en-2-yl) amino) -4-oxo-1, 4-dihydropyridine-2, 5-dicarboxamide
Tert-butyl (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxopyridin-1 (4H) -yl) (3-methylbutan-3-en-2-yl) carbamate (0.780 g,1.14 mmol) was dissolved in DCM (6.0 mL) and a solution of hydrogen chloride in 1, 4-dioxane (4.00 mol/L,1.42mL,5.69 mmol) was added. The reaction was stirred at ambient temperature for 21 hours. The reaction was concentrated and purified by flash chromatography (hexanes: etOAc) to give 3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -N5- (2, 4-difluorobenzyl) -1- ((3-methylbut-3-en-2-yl) amino) -4-oxo-1, 4-dihydropyridine-2, 5-dicarboxamide. ES/MS 551.183 (M+H+).
Step 3: preparation of 3- ((S) -but-3-en-2-yl) -N- (2, 4-difluorobenzyl) -5-hydroxy-1- (3-methylbut-3- Alkene-2-yl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -N5- (2, 4-difluorobenzyl) -1- ((3-methylbut-3-en-2-yl) amino) -4-oxo-1, 4-dihydropyridine-2, 5-dicarboxamide (0.630 g,1.14 mmol) was dissolved in 1:1DCE: ACN (3.0 mL). To this solution was added paraformaldehyde (309 mg,3.43 mmol), acetic acid (0.463 mL,8.01 mmol) and trifluoroacetic acid (0.013 mL,5.72 mmol). The reaction was heated to 90 ℃ overnight (-12 hours). The reaction mixture was cooled to ambient temperature and concentrated mostly by rotary evaporation. The residue obtained is treated with saturated NaH CO 3 The aqueous solution was quenched and extracted with EtOAc (3X 10 mL). The organic layers were combined and washed with brine (1×20 mL), then dried over sodium sulfate, then concentrated by rotary evaporation. The residue was purified by flash chromatography (hexanes: etOAc) to give 3- ((S) -but-3-en-2-yl) -N- (2, 4-difluorobenzyl) -5-hydroxy-1- (3-methylbut-3-en-2-yl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide. ES/MS 473.252 (M+H+).
Step 4: preparation of 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -N- (2, 4-difluorobenzyl) -1- (3-methyl) But-3-en-2-yl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
3- ((S) -but-3-en-2-yl) -N- (2, 4-difluorobenzyl) -5-hydroxy-1- (3-methylbut-3-en-2-yl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (117 mg,0.247 mmol) was dissolved in DMF (2.5 mL). Potassium carbonate (0.205 g,0.00148 mol) was added followed by benzyl bromide (0.106 g, 0.611 mmol). The reaction was heated at 90℃for 4 hours. The mixture was cooled to ambient temperature and diluted with water (5 mL). The reaction was extracted with EtOAc (3X 5 mL). The organic layers were combined and washed with brine (2×10 mL), dried over sodium sulfate and concentrated by rotary evaporation. The residue was purified by flash chromatography (hexanes: etOAc) to give 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -N- (2, 4-difluorobenzyl) -1- (3-methylbut-3-en-2-yl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide. ES/MS 563.183 (M+H+).
Step 5: preparation of (1S, 2R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2,3, 5-trimethyl-7, 9-dione Oxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -N- (2, 4-difluorobenzyl) -1- (3-methylbut-3-en-2-yl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (0.117 g,0.207 mmol) was dissolved in DCE (6 mL) and the 2 nd generation Hoveyda-Grubbs catalyst (51.9 mg,0.0828 mmol) was added. The mixture was sprayed three times with Ar and then heated to 90℃overnight (. About.12 hours). The mixture was cooled to ambient temperature and concentrated by rotary evaporation. Purification by flash chromatography (hexane: etOAc) afforded (1S, 2R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2,3, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide. ES/MS 535.102 (M+H+).
Step 6: preparation of (1S, 2R, 5S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 3, 5-trimethyl-7, 9-dioxo- 2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2,3, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (3.10 mg, 5.80. Mu. Mol) was dissolved in DMF (500. Mu.L) and lithium chloride (7.00 mg, 165. Mu. Mol) was added. The reaction was heated at 90 ℃ overnight. The mixture was cooled to ambient temperature, filtered and purified by reverse phase HPLC (water with 0.1% tfa: ACN) to give (1 s,2r,5 s) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 3, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide. ES/MS 445.213 (M+H+). 1H NMR (400 MHz, acetonitrile-d 3) δ10.17 (s, 1H), 8.34 (s, 1H), 7.41 (q, J=9.0, 8.3Hz, 1H), 7.09-6.80 (m, 2H), 5.51 (d, J=2.3 Hz, 1H), 5.15 (d, J=7.1 Hz, 1H), 5.07 (d, J=14.5 Hz, 1H), 4.72-4.40 (m, 3H), 3.83 (d, J=6.9 Hz, 1H), 1.86-1.68 (m, 3H), 1.28 (dd, J=15.0, 7.0Hz, 6H). 19F NMR (376 MHz, acetonitrile-d 3) delta-114.02 (p, J=7.8, 7.4 Hz), -115.37-118.64 (m).
Example 63: preparation of (1S, 2R, 5S) -N- (3-chloro-2, 4-difluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9- Dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (C63)
Step 1: preparation((1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-) 1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carbonyl) (2, 4, 6-trifluorobenzyl) carbamic acid Tert-butyl ester
(1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (837 mg,1.55 mmol) was dissolved in toluene (12 mL) at room temperature. DMAP (570 mg,4.66 mmol) and (Boc) 2O (1.355 g,6.22 mmol) were added sequentially. The reaction mixture was heated at 110 ℃ with stirring for 2 hours and then concentrated to dryness. The residue was purified on a silica gel column with 0% -100% etoac/Hex to give the product. MS (m/z): 639.2[ M+H ] +.
Step 2: preparation of (1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxylic acid
Tert-butyl ((1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carbonyl) (2, 4, 6-trifluorobenzyl) carbamate (635 mg,0.994 mmol) was dissolved in MeOH (12 mL) followed by water (6 mL). NaOH (1M, 3.5mL,3.4 mmol) was then added dropwise. The resulting reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was then diluted with water (20 mL) and acidified with 1N HCl to ph=3. EtOAc (50 mL) was added for extraction. The organic phase was separated and washed with brine (50 mL). The organic phase was separated and dried over Na2SO 4. Filtration and concentration gave the crude product which was purified by reverse phase prep HPLC with 0% to 100% acetonitrile in water containing 0.1% tfa to give the desired product. MS (m/z): 396.1[ M+H ] +.
Step 3: preparation of (1S, 2R, 5S) -8- (benzyloxy) -N- (3-chloro-2, 4-difluorobenzyl) -2, 5-dimethyl-7, 9- Dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxylic acid (13 mg,0.0329 mmol) was dissolved in DMF (1 mL) at room temperature. DIEA (17 mg,0.132 mmol) was added under an argon atmosphere. The resulting reaction mixture was cooled to 0 ℃. HATU (18.8 mg,0.0493 mmol) was then added. The resulting reaction mixture was then warmed to room temperature and stirred at room temperature for 1 hour. To the reaction mixture was added a solution of (3-chloro-2, 4-difluorophenyl) methylamine (8.76 mg,0.0493 mmol) in DMF (0.5 mL). The reaction mixture was then stirred at room temperature for 17 hours. The reaction mixture was diluted with EtOAc (10 mL) and treated with a mixture of saturated aqueous NH4Cl (10 mL) and water (10 mL). The organic phase was then washed with water (10 mL) followed by saturated brine (10 mL). The organic phase was then separated and concentrated. The residue was purified on a silica gel column with 0% -100% etoac/Hex to give the product. MS (m/z): 555.2[ M+H ] +.
Step 4: preparation of (1S, 2R, 5S) -N- (3-chloro-2, 4-difluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo Substituted-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -N- (3-chloro-2, 4-difluorobenzyl) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] at room temperature][1,2,5]Triazacyclononane-10-carboxamide (17 mg,0.0306 mmol) was dissolved in toluene (2 mL). TFA (2 mL) was carefully added with stirring. The resulting reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was then concentrated to dryness. The residue was dissolved in MeOH and taken up in 0.1% TFA in 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilizing to obtain the final product. MS (m/z): 465.2[ M+H ]]+。1H NMR(400MHz,CD3CN)δ10.26(s,1H),8.39(s,1H),7.38(td,J=8.4,6.1Hz,1H),7.10(td,J=8.7,1.8Hz,1H),5.66(dt,J=11.4,2.4Hz,1H),5.46–5.31(m,2H),5.02(d,J=14.4Hz,1H),4.64(m,2H),4.58(d,J=14.4Hz,1H),3.84(tq,J=6.7,3.6Hz,1H),1.35(dd,J=7.1,2.3Hz,6H)。
Example 64: preparation of (1S, 2R, 5R) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopisPyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C64)
(1S, 2R, 5R) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide is prepared in analogy to example 24 except that (R) -but-3-en-2-amine hydrochloride is used instead of (S) -but-3-en-2-amine hydrochloride in step 4. MS (m/z): 449.2[ M+H ] +.1H NMR (400 MHz, CD3 CN) δ10.31 (s, 1H), 8.39 (s, 1H), 6.93-6.81 (m, 2H), 5.80 (ddd, J=11.7, 2.7,1.9Hz, 1H), 5.45 (ddd, J=11.7, 4.1,2.4Hz, 1H), 4.82-4.55 (m, 4H), 4.32 (dtt, J=7.4, 5.0,2.5Hz, 1H), 4.01 (ddt, J=6.8, 4.5,2.2Hz, 1H), 1.79 (d, J=7.5 Hz, 3H), 1.40 (d, J=7.0 Hz, 3H).
Example 65: preparation of (1S, 2R, 5R) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C65)
(1S, 2R, 5R) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 64][1,2,5]Triazacyclononane-10-carboxamide (25 mg,0.0558 mmol) was dissolved in MeOH (10 mL). Pd/C (10%) (12 mg) was added. With H at room temperature 2 The balloon was subjected to hydrogenolysis for 7 hours. The reaction mixture was filtered through celite. The filtrate was collected and concentrated to dryness. The residue was dissolved in MeOH and taken up in 0.1% TFA in 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilizing to obtain the final product. MS (m/z): 451.2[ M+H ]]。1H NMR(400MHz,CD3CN)δ10.30(s,1H),8.38(s,1H),6.87(t,J=8.5Hz,2H),4.75–4.51(m,4H),3.60–3.42(m,1H),3.00(dq,J=12.2,6.4Hz,1H),1.59–1.76(m,4H),1.73(d,J=7.0Hz,3H),1.30(d,J=6.5Hz,3H)。
Example 66: preparation of (1R, 2S, 5S) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C66)
(1R, 2S, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 24][1,2,5]Triazacyclononane-10-carboxamide (16 mg,0.0297 mmol) was dissolved in MeOH (10 mL). Pd-C (10%) (12 mg) was added. With H at room temperature 2 The balloon was subjected to hydrogenolysis for 7 hours. The reaction mixture was filtered through celite. The filtrate was collected and concentrated to dryness. The residue was dissolved in MeOH and taken up in 0.1% TFA in 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilizing to obtain the final product. MS (m/z): 451.2[ M+H ]]。1H NMR(400MHz,CD 3 CN)δ10.30(s,1H),8.38(s,1H),6.87(t,J=8.5Hz,2H),4.75–4.51(m,4H),3.60–3.42(m,1H),3.00(dq,J=12.2,6.4Hz,1H),1.59–1.76(m,4H),1.73(d,J=7.0Hz,3H),1.30(d,J=6.5Hz,3H)。
Example 67: preparation of (1S, 2R, 5R) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo Substituted-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (C67)
Step 1: preparation of (1R, 2S, 5R) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-7, 9-dioxo Substituted-2, 5,7, 9-tetrahydro-1, 6-methanopyridinesAnd [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (A1), (1R, 2R, 5R) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (B1) and (1S, 2R, 5R) -8- (benzyloxy) Phenyl) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1, 2-b][1,2,5]triazacyclononane-10-carboxamide (C1)
5- (benzyloxy) -3- ((R) -but-3-en-2-yl) -1- (but-3-en-2-yl) -N- (2, 4-difluorobenzyl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (390 mg,0.711 mmol) prepared at room temperature in analogy to example 29 except that (R) -but-3-en-2-amine was used instead of (S) -but-3-en-2-amine was dissolved in dichloromethane (60 mL). Argon was bubbled through the reaction solution for 20 minutes. HG-M720 catalyst (44.5 mg,0.071 mmol) was then added with stirring. Purging with argon was continued for 10 minutes. The reaction mixture was then heated under argon atmosphere with a reflux condenser for 24 hours. The resulting reaction mixture was then concentrated to dryness. The crude material was purified on a silica gel column with 0% -100% etoac/Hex to give three diastereomers. MS (m/z): 521.1[ M+H ] +.
Step 2: preparation of (1S, 2R, 5R) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R, 5R) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] at room temperature][1,2,5]Triazacyclononane-10-carboxamide (10 mg,0.019 mmol) was dissolved in EtOH (3 mL) and 10% Pd-C (4 mg,0.0038 mmol) was added. With H at room temperature 2 The balloon was subjected to hydrogenolysis for 7 hours. The reaction mixture was filtered through a celite pad. The filtrate was collected and concentrated to dryness. The residue was dissolved in MeOH and taken up in 50% -100% CH with 0.1% TFA 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. MS (m/z): 433.2[ M+H ]]+。1H NMR (400 MHz, acetonitrile-d 3) delta 10.33 (d, j=30.5 hz, 1H), 8.41 (d, j=19.1 hz, 1H), 7.43 (td, j=9.2, 8.8,6.5hz, 1H), 7.06-6.85 (m, 2H), 4.71 (d, j=14.9 hz, 1H), 4.60 (d, j=5.8 hz, 2H), 4.56 (s, 1H), 3.51 (ddt, j=11.2, 7.1,3.5hz, 1H), 3.31-3.01 (m, 1H), 2.07-1.99 (m, 1H), 1.93-1.66 (m, 6H), 1.30 (p, j=6.6 hz, 3H).
Example 68: preparation of (1R, 2R, 5R) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo Substituted-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C68)
(1R, 2R, 5R) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 67][1,2,5]Triazacyclononane-10-carboxamide (21 mg,0.04 mmol) dissolved in CH 3 CN (2 mL) was added MgBr 2 (22.3 mg,0.22 mmol) and stirred at 50℃for 2 hours. The reaction mixture was quenched with water (1 mL) to form a clear solution, filtered and the residue was dissolved in MeOH and taken up in 50% -100% CH 3 The CN aqueous solution was purified by reverse phase prep HPLC to give the desired product. MS (m/z): 431.2[ M+H ]]+.1H NMR (400 MHz, acetonitrile-d 3) δ10.28 (s, 1H), 8.45 (s, 1H), 7.44 (q, J=9.1, 8.4Hz, 1H), 6.98 (tt, J=10.9, 3.1Hz, 2H), 5.69-5.54 (m, 1H), 5.47-5.26 (m, 2H), 4.94 (d, J=14.3 Hz, 1H), 4.73 (d, J=14.4 Hz, 1H), 4.61 (d, J=5.7 Hz, 3H), 1.36 (d, J=7.2 Hz, 3H), 1.03 (d, J=7.3 Hz, 3H).
Example 69: preparation of (1S, 2R, 5R) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo Substituted-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C69)
Will depend on the implementation(1S, 2R, 5R) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared in example 67 ][1,2,5]Triazacyclononane-10-carboxamide (8 mg,0.015 mmol) dissolved in CH 3 To CN (2 mL), mgBr2 (6.2 mg,0.034 mmol) was added and stirred at 50℃for 2 hours. The reaction mixture was quenched with water (1 mL) to form a clear solution, filtered and the residue was dissolved in MeOH, and taken up in 50% -100% CH 3 The CN aqueous solution was purified by reverse phase prep HPLC to give the desired product. MS (m/z): 431.1[ M+H ]]+.1H NMR (400 MHz, acetonitrile-d 3) δ10.31 (s, 1H), 8.44 (d, J=9.2 Hz, 1H), 7.44 (H, J=6.6 Hz, 1H), 6.98 (dt, J=13.7, 4.9Hz, 2H), 5.89-5.72 (m, 1H), 5.55-5.33 (m, 1H), 4.83-4.67 (m, 2H), 4.66-4.52 (m, 2H), 4.33 (ddd, J=8.8, 5.6,3.2Hz, 1H), 4.23-3.90 (m, 1H), 1.80 (d, J=7.5 Hz, 3H), 1.41 (d, J=7.0 Hz, 3H).
Example 70: preparation of (1S, 2R) -8-hydroxy-2-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5, 7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C70)
(1S, 2R) -8-hydroxy-2-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide was prepared in analogy to example 24 except that allyl ammonium chloride was used instead of (S) -but-3-en-2-amine hydrochloride in step 4. ES/MS (m/z): 435.199[ M+H ] +,1H NMR (400 MHz, acetonitrile-d 3) δ10.19 (s, 1H), 8.37 (s, 1H), 6.85 (t, J=8.6 Hz, 2H), 5.74 (dq, J=11.7, 2.8Hz, 1H), 5.43 (ddt, J=11.8, 3.6,2.2Hz, 1H), 4.95 (dq, J=18.4, 2.9Hz, 1H), 4.84 (d, J=14.4 Hz, 1H), 4.70 (d, J=14.3 Hz, 1H), 4.64-4.57 (m, 2H), 3.92-3.74 (m, 2H), 1.36 (d, J=6.8 Hz, 3H).
Example 71: preparation of (1S, 2R) -8-hydroxy-2-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3, 4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (C71)
Addition of (1S, 13R) -6-hydroxy-13-methyl-5, 8-dioxo-N- [ (2, 4, 6-trifluorophenyl) methyl ] prepared according to example 70 to a vial]-1,2, 9-triazatricyclo [7.4.1.02,7 ]]Tetradecane-3,6,11-triene-4-carboxamide (37.6 mg,0.086mmol,1.0 eq.) platinum (IV) oxide (1 mg, 4.3. Mu. Mol,5 mol%) and ethyl acetate (1.5 mL). The vial was then filled with hydrogen balloon and hydrogen gas was bubbled through the reaction mixture for 5 minutes. The reaction was left under hydrogen atmosphere for 39 hours, then the grey suspension was filtered and the volatiles were removed in vacuo, and the resulting residue was purified by preparative HPLC (0% -100% ch with 0.1% tfa modifier 3 CN/H 2 O) to give (1S, 2R) -8-hydroxy-2-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide. ES/MS (m/z): 437.168[ M+H ]]+.1H NMR (400 MHz, chloroform-d) δ10.20 (s, 1H), 8.54 (s, 1H), 6.66 (t, J=8.1 Hz, 2H), 4.76-4.58 (m, 3H), 4.49-4.36 (m, 2H), 3.26-3.16 (m, 1H), 3.05 (dt, J=13.4, 6.5Hz, 1H), 1.98 (q, J=5.5 Hz, 2H), 1.74 (q, J=5.0 Hz, 2H), 1.35 (d, J=6.8 Hz, 3H).
Example 72: preparation of (1S, 2R, 5S) -2-ethyl-8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluoro) Benzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C72)
Preparation of 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -4, 6-dioxo-1- (pent-1-en-3-yl) -N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
To a solution of (S) -5- (benzyloxy) -3- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (500 mg,0.976 mmol) prepared according to example 46 in THF was added 1-ethyl allyl acetate (625 mg,4.88 mmol), 1, 8-diazabicyclo [5.4.0] undec-7-ene (1.5 g,9.76 mmol) and tetrakis (triphenylphosphine) palladium (0) (225 mg,0.195 mmol). The reaction mixture was heated at 65 ℃. After completion of the reaction, the reaction mixture was concentrated and the residue was purified by silica gel chromatography eluting with 0% -100% hexanes/EtOAc. MS (m/z) 581.19[ M+H ] +.
Preparation of (1S, 2R, 5S) -8- (benzyloxy) -2-ethyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (A) and (1S, 2S, 5S) -8- (benzyloxy) -2-ethyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetraf luorobenzyl) Hydrogen-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (B)
A solution of (S) -5- (benzyloxy) -3- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (60 mg,0.103 mmol) in DCE (8 mL) was degassed with Ar for 5 min. To the mixture was added a catalytic Hoverda-Grubbs II catalyst M720 (6.5 mg,0.013 mmol) and the mixture was sparged with Ar for 10 minutes. The reaction mixture was then stirred at 80 ℃ overnight. The reaction mixture was concentrated and the residue was purified by silica gel chromatography eluting with 0% -100% hexanes/EtOAc to give (1 s,2r,5 s) -8- (benzyloxy) -2-ethyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-B ] [1,2,5] triazacyclononane-10-carboxamide (a) as the main product and (1 s,2s,5 s) -8- (benzyloxy) -2-ethyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-B ] [1,2,5] triazacyclononane-10-carboxamide (B) as the minor product.
(1 s,2r,5 s) -8- (benzyloxy) -2-ethyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (a): MS (m/z) 553.06[ M+H ] +.
(1S, 2S, 5S) -8- (benzyloxy) -2-ethyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (B) MS (m/z) 553.05[ M+H ]] +
Preparation of (1S, 2R, 5S) -2-ethyl-8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -2-ethyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]A solution of triazacyclononane-10-carboxamide (15 mg,0.0258 mmol) in DCM (1 mL) and TFA (1 mL) was stirred overnight at room temperature. The mixture was then concentrated and purified by reverse phase HPLC with 5% -100% aqueous acetonitrile to give the title product. MS (m/z) 462.21[ M+H ]] + .1H NMR (400 MHz, methanol-d 4) delta 8.43 (s, 1H), 7.01-6.84 (m, 2H), 5.77 (dt, J=11.6, 2.4Hz, 1H), 5.58-5.46 (m, 1H), 5.42 (dt, J=7.5, 2.8Hz, 1H), 5.08 (d, J=14.4 Hz, 1H), 4.75-4.56 (m, 3H), 3.76 (dp, J=6.7, 3.4Hz, 1H), 1.88-1.59 (m, 2H), 1.39 (d, J=7.3 Hz, 3H), 1.08 (t, J=7.3 Hz, 3H).
Example 73: preparation of (1S, 2R, 5S) -2-ethyl-8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluoro) Benzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C73)
To (1S, 2R, 5S) -2-ethyl-8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 72][1,2,5]To a solution of triazacyclononane-10-carboxamide (5 mg,0.01 mmol) in EtOH (2 ml) was added PtO 2 (1 mg). The reaction mixture was taken up at room temperature at H 2 Stirring under balloon2 hours. The reaction mixture was filtered through celite and concentrated. The residue was purified by reverse phase HPLC with 5% -100% acn/H2O containing 0.1% tfa to give the title compound. MS (m/z) 465.24[ M+H ]] + .1H NMR (400 MHz, methanol-d 4) delta 8.44 (s, 1H), 6.97-6.87 (m, 2H), 4.80-4.52 (m, 5H), 3.24 (t, J=7.1 Hz, 1H), 2.04 (dt, J=14.3, 6.9Hz, 1H), 1.97-1.66 (m, 3H), 1.63-1.43 (m, 2H), 1.31 (d, J=6.7 Hz, 3H), 1.19 (t, J=7.3 Hz, 3H).
Example 74: preparation of (1S, 2R, 5S) -8-hydroxy-2-isopropyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trio Fluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C74)
Preparation of (1S, 2R, 5S) -8- (benzyloxy) -2-isopropyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -2-isopropyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide is prepared similarly to (1S, 2R, 5S) -8- (benzyloxy) -2-ethyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide in example 72 except (4-methylpent-1-en-3-yl) carbonate is used instead of pent-1-en-3-yl acetate. MS (m/z) 567.08[ M+H ] +.
Preparation of (1S, 2R, 5S) -8-hydroxy-2-isopropyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) scheme 2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -2-isopropyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanoneMethylpyrido [1,2-b ]][1,2,5]A reaction mixture of triazacyclononane-10-carboxamide (10 mg,0.017 mmol) and lithium chloride (7.5 mg,0.17 mmol) in DMF (1 mL) was heated at 100deg.C overnight. The reaction mixture was filtered and the solution purified by reverse phase HPLC eluting with 5% to 100% acetonitrile/water to give the title compound. MS (m/z) 477.21[ M+H ] ] + .1H NMR (400 MHz, methanol-d 4) delta 8.42 (s, 1H), 6.95-6.86 (m, 2H), 5.83 (dt, J=11.7, 2.4Hz, 1H), 5.56 (dt, J=11.8, 3.1Hz, 1H), 5.42 (dtd, J=9.9, 7.0,4.1Hz, 1H), 5.05 (d, J=14.4 Hz, 1H), 4.75-4.62 (m, 3H), 3.67 (p, J=3.3 Hz, 1H), 2.00 (dtq, J=10.4, 7.0,3.9Hz, 1H), 1.39 (d, J=7.3 Hz, 3H), 1.17 (d, J=6.7 Hz, 3H), 0.92 (d, J=7.0 Hz, 3H).
Example 75: preparation of (1S, 2S, 5S) -8-hydroxy-2-isopropyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trio Fluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxylic acid Amine (C75)
(1S, 2S, 5S) -8-hydroxy-2-isopropyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide is synthesized in analogy to example 73, except that (1S, 2R, 5S) -8-hydroxy-2-isopropyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] is used][1,2,5]Triazacyclononane-10-carboxamide replaces (1S, 2R, 5S) -2-ethyl-8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 479.21[ M+H ] ] + .1H NMR (400 MHz, methanol-d 4) delta 8.43 (s, 1H), 6.91 (t, J=8.4 Hz, 2H), 4.82-4.53 (m, 5H), 2.89 (d, J=10.0 Hz, 1H), 2.11-1.98 (m, 2H), 1.94 (dp, J=16.1, 4.4,3.9Hz, 1H), 1.73 (dt, J=14.7, 10.2Hz, 1H), 1.44 (ddd, J=14.8, 11.0,3.1Hz, 1H), 1.30 (dd, J=10.4, 6.6Hz, 6H), 0.97 (s, 3H).
Example 76: preparation of the product (1S,2S, 5S) -2- (fluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-) Trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C76)
Step 1: synthesis of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) Radical) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid
3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid methyl ester (5.9 g,10.5 mmol) was dissolved in 100mL MeOH and 20mL water, lithium hydroxide (1.26 g,52.5 mmol) was added at room temperature, then heated to 60℃overnight, then heated to 70℃for 8 hours. The reaction was cooled to 0 ℃, 2N HCl was added to adjust the pH to 3. The reaction crude product was concentrated. 100mL of EtOAc was added. The precipitate was filtered and washed with water (30 ml 2×). The solid was dried in vacuo to give 4.82g of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid.
Step 2: synthesis of (S) -3- (benzyloxy) -2- (but-3-en-2-ylcarbamoyl) -4-oxo-5- ((2, 4, 6-) Trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) carbamic acid tert-butyl ester
3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid (5.2 g,9.5 mmol) and (2S) -but-3-en-2-amine hydrochloride (1.23 g,11.4 mmol) were dissolved in anhydrous DMF (500 mL) and cooled to 0deg.C. HATU (4.3 g,11.4 mmol) and 1-hydroxy-7-azabenzotriazole (3838 mg,2.85 mmol) were added followed by DIEA (4.96 ml,28.5 mmol). The reaction was maintained at 0 ℃. After 10 minutes, the reaction was completed. The reaction mixture was poured into ice water and extracted with EtOAc (400 ml 2×). The organic layer was concentrated and passed through silica gelColumn purification eluting with EtOAc/hexanes (20% -70%) afforded tert-butyl (S) - (3- (benzyloxy) -2- (but-3-en-2-ylcarbamoyl) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) carbamate. MS (m/z) 601.2[ M+H] +
Step 3: synthesis of (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2), 4, 6-Trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) ((S) -1-hydroxybut-3-en-2-yl) carbamic acid tert-butyl ester
Tert-butyl (S) - (3- (benzyloxy) -2- (but-3-en-2-ylcarbamoyl) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) carbamate (5.09 g,8.48 mmol) was dissolved in 500mL anhydrous MeCN, tetrabutylammonium bromide (5.46 g,17 mmol) was added followed by (R, R) -DACH naphtyltriost ligand (264 mg,1.02 mmol) and tris (dibenzylideneacetone) dipalladium (439 mg, 0.284 mmol). Ar was purged for 10 minutes, and then butadiene monoxide (1.71 mL,21.2 mmol) was added dropwise to the reaction. The reaction was stirred at room temperature for 3 hours. The crude reaction product was concentrated and purified by column on silica gel eluting with EtOAc/hexanes (30% -70%) to give tert-butyl (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) ((S) -1-hydroxybut-3-en-2-yl) carbamate. MS (m/z) 671.3[ M+H ]] +
Step 4: synthesis of (S) -2- ((3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-o- 5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) (tert-butoxycarbonyl) amino) but-3-en-1-ylethylene Acid esters
Tert-butyl (3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) ((S) -1-hydroxybut-3-en-2-yl) carbamate (1.54 g,2.29 mmol) was dissolved in pyridine (30 mL) and acetic anhydride (3.25 mL,34.4 mmol) and 1-hydroxy-7-azabenzotriazole (578 mg,2.85 mmol) were added followed by DMAP (560 mg,4.59 mmol). The reaction was heated at 50℃for 4 days. The crude reaction product was concentrated and purified by column on silica gel Elution with EtOAc/hexanes (40% -70%) afforded (S) -2- ((3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) (tert-butoxycarbonyl) amino) but-3-en-1-yl acetate. MS (m/z) 713.4[ M+H ]] +
Step 5: synthesis of (S) -2- ((3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-o- 5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) amino) but-3-en-1-yl acetate
(S) -2- ((3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) (tert-butoxycarbonyl) amino) but-3-en-1-yl acetate (1.13 g,2.29 mmol) was dissolved in dichloromethane (3 mL) and a solution of 4N HCl in dioxane (4 mL) was added at room temperature. The reaction was stirred at room temperature for one hour. The reaction was completed. The crude reaction was concentrated and purified by column on silica gel eluting with EtOAc/hexanes (40% -80%) to give (S) -2- ((3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) amino) but-3-en-1-yl acetate. MS (m/z) 613.3[ M+H ] ] +
Step 6: synthesis of (S) -2- (3- ((S) -but-3-en-2-yl) -5-hydroxy-4, 6-dioxo-7- ((2, 4, 6-trio) Fluorobenzyl) carbamoyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazin-1-yl) but-3-en-1-yl Acetic acid esters
(S) -2- ((3- (benzyloxy) -2- (((S) -but-3-en-2-yl) carbamoyl) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) pyridin-1 (4H) -yl) amino) but-3-en-1-yl acetate (405 mg,0.66 mmol) was dissolved in 1, 2-dichloroethane (17 mL) and acetonitrile (17 mL), paraformaldehyde (60 mg,1.98 mmol) was added. Acetic acid (0.265 mL,4.63 mmol) and TFA (0.803 mL,3.31 mmol) were then added simultaneously to the reaction. The reaction was heated at 90℃for 20 hours. The crude reaction was concentrated and purified by column on silica gel eluting with EtOAc/hexanes (40% -100%) to give (S) -2- (3- ((S) -but-3-en-2-yl) -5-hydroxy-4, 6-dioxo-7- ((2, 4, 6-trifluorobenzyl)Carbamoyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazin-1-yl) but-3-en-1-yl acetate. MS (m/z) 535.2[ M+H ]] +
Step 7: synthesis of (S) -2- (5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -4, 6-dioxo-7- ((2, 4), 6-trifluorobenzyl) carbamoyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazin-1-yl) but-3-ene 1-yl acetate
(S) -2- (3- ((S) -but-3-en-2-yl) -5-hydroxy-4, 6-dioxo-7- ((2, 4, 6-trifluorobenzyl) carbamoyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazin-1-yl) but-3-en-1-yl acetate (163 mg,0.3 mmol) was dissolved in DMF (3 mL) and K was added 2 CO 3 (208 mg,1.5 mmol) followed by bromomethylbenzene (0.072 mL,0.6 mmol). The reaction was stirred at room temperature overnight. The crude reaction was taken up with EtOAc and saturated NaHCO 3 And (5) extracting the solution. The organic layer was concentrated and purified by column on silica gel eluting with EtOAc/hexanes (40% -100%) to give (S) -2- (5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -4, 6-dioxo-7- ((2, 4, 6-trifluorobenzyl) carbamoyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazin-1-yl) but-3-en-1-yl acetate. MS (m/z) 625.3[ M+H ]] +
Step 8: synthesis of ((1S, 2S, 5S) -8- (benzyloxy) -5-methyl-7, 9-dioxo-10- ((2, 4, 6-trifluorobenzyl) Radical) carbamoyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-2- Methyl radical) acetate
(S) -2- (5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -4, 6-dioxo-7- ((2, 4, 6-trifluorobenzyl) carbamoyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazin-1-yl) but-3-en-1-yl acetate (210 mg,0.336 mmol) was dissolved in 1, 2-dichloroethane (17 mL) and the 2 nd generation Hoveyda-Grubbs catalyst (42 mg,0.067 mmol) was added. The reaction was heated at 80℃for 6 hours. The crude reaction was concentrated and purified by column on silica gel eluting with EtOAc/hexanes (40% -100%) to give ((1 s,2s,5 s) -8- (benzyloxy) -5-methyl-7, 9-dioxo-10- ((2, 4, 6-trifluorobenzyl) carbamoyl) -2,5,7, 9-tetrahydro-1, 6-methano) Pyrido [1,2-b][1,2,5]Methyl triazacyclononan-2-yl) acetate. MS (m/z) 597.3[ M+H ]] +
Step 9: synthesis of (1S, 2S, 5S) -8- (benzyloxy) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxylic acid Amines
((1S, 2S, 5S) -8- (benzyloxy) -5-methyl-7, 9-dioxo-10- ((2, 4, 6-trifluorobenzyl) carbamoyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Methyl triazacyclononan-2-yl acetate (135 mg,0.226 mmol) was dissolved in MeOH (3 mL) and K was added 2 CO 3 (93.8 mg,0.679 mmol). The reaction was stirred at room temperature for 15 minutes. EtOAc was added to the crude reaction. Transferred to a separate funnel and water was added to wash the organic layer twice. The organic layer was concentrated and purified by silica column eluting with EtOAc/hexanes (60% -100%) to give (1 s,2s,5 s) -8- (benzyloxy) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1, 2-b)][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 555.3[ M+H ]] +
Step 10: synthesis of (1S, 2S, 5S) -8- (benzyloxy) -2- (fluoromethyl) -5-methyl-7, 9-dioxo-N- (2, 4), 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10-carboxylic acid Amines
(1S, 2S, 5S) -8- (benzyloxy) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (24 mg,0.05 mmol) was dissolved in DCM (1.5 mL) and cooled in an ice bath. Deoxyfluor (50% toluene solution, 2.7M,0.39 mL) was added. The reaction was stirred at 0 ℃ and then warmed to room temperature for 2 hours. Addition of saturated NaHCO 3 The solution was used to quench the reaction. Extraction using DCM. The organic layer was concentrated and purified by preparative HPLC eluting with 10% to 60% acetonitrile (0.1% tfa) in water (0.1% tfa) to give (1 s,2s,5 s) -8- (benzyloxy) -2- (fluoromethyl) -5-methyl-7, 9-di-oxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 557.2[ M+H ]] +
Step 11: synthesis of (1S, 2S, 5S) -2- (fluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trio Fluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2S, 5S) -8- (benzyloxy) -2- (fluoromethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10-carboxamide (15 mg,0.027 mmol) was dissolved in toluene (0.5 mL) and TFA (0.5 mL) was added. The reaction was stirred at room temperature for 4 hours. Concentration of the reaction and purification via preparative HPLC eluting with 10% -60% acetonitrile (0.1% TFA) in water (0.1% TFA) afforded (1S, 2S, 5S) -2- (fluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1, 2-b)][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 467.2[ M+H] +1 H NMR (400 MHz, acetonitrile-d) 3 )δ10.21(s,1H),8.46(d,J=1.1Hz,1H),6.87(t,J=8.5Hz,2H),5.82(dt,J=11.7,2.8Hz,1H),5.39(ddt,J=14.7,12.0,3.4Hz,2H),4.95(d,J=14.5Hz,1H),4.70–4.59(m,4H),4.54–4.49(m,1H),4.09(ddd,J=18.4,5.9,3.0Hz,1H),1.35(d,J=7.3Hz,3H)。
Example 77: preparation of (1S, 2S, 5S) -2- (fluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4,6- Trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-A Amide (C77)
(1S, 2S, 5S) -2- (fluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 76][1,2,5]Triazacyclononane-10-carboxamide (6 mg, 0.0)13 mmol) was dissolved in EtOH (5 mL) and EtOAc (5 mL). 10% Pd/C (3 mg) was added and a hydrogen balloon was applied. The reaction was stirred at room temperature for 2 hours. The reaction was filtered through celite, the filtrate was concentrated and purified by preparative HPLC eluting with 10% to 60% acetonitrile (0.1% tfa) in water (0.1% tfa) to give (1 s,2s,5 s) -2- (fluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] ][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 469.2[ M+H] + .1H NMR (400 MHz, acetonitrile-d 3) δ10.31 (s, 1H), 8.40 (s, 1H), 7.01-6.80 (m, 2H), 4.77-4.68 (m, 1H), 4.65-4.56 (m, 4H), 4.47 (dd, J=9.9, 5.4Hz, 1H), 3.67-3.45 (m, 2H), 2.06 (dt, J=13.2, 7.0Hz, 1H), 1.84 (ddd, J=15.5, 7.9,3.9Hz, 1H), 1.75-1.60 (m, 2H), 1.26 (d, J=6.7 Hz, 3H).
Example 78: preparation of (1S, 2S, 5S) -2- (difluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4), 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10- Formamide (C78)
Step 1: synthesis of ((1S, 2S, 5S) -8-hydroxy-5-methyl-7, 9-dioxo-10- ((2, 4, 6-trifluorobenzyl) ammonia Carbamoyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononan-2-yl Acetic acid methyl ester
((1S, 2S, 5S) -8- (benzyloxy) -5-methyl-7, 9-dioxo-10- ((2, 4, 6-trifluorobenzyl) carbamoyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Methyl triazacyclononan-2-yl acetate (48 mg,0.08 mmol) was dissolved in EtOH (5 mL) and EtOAc (5 mL), 10% Pd/C (16 mg) was added and a hydrogen balloon was applied. The reaction was stirred at room temperature for 2 hours. The reaction was filtered through celite and the filtrate was concentrated to give ((1S, 2S, 5S) -8-hydroxy-5-methyl-7, 9-dioxo-10- ((2, 4, 6-trifluorobenzyl) carbamoyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1, 2-b][1,2,5]Methyl triazacyclononan-2-yl) acetate, which is used directly in the next step. MS (m/z) 509.2[ M+H ]] +
Step 2: synthesis of ((1S, 2S, 5S) -8- (benzyloxy) -5-methyl-7, 9-dioxo-10- ((2, 4, 6-trifluorobenzyl) Radical) carbamoyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane (T-N-phenylmethane) 2-yl) acetic acid methyl ester
((1S, 2S, 5S) -8-hydroxy-5-methyl-7, 9-dioxo-10- ((2, 4, 6-trifluorobenzyl) carbamoyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]The crude product of methyl triazacyclononan-2-yl acetate was dissolved in DMF (3 mL) and K was added 2 CO 3 (22 mg,0.16 mmol) followed by BnBr (0.014 mL,0.12 mmol). The reaction was stirred at room temperature for 2 hours. The reaction was extracted with EtOAc/saturated NaHCO3, the organic layer was concentrated, purified by column on silica gel, eluting with EtOAc/hexanes (60% -100%) to give ((1 s,2s,5 s) -8- (benzyloxy) -5-methyl-7, 9-dioxo-10- ((2, 4, 6-trifluorobenzyl) carbamoyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1, 2-b)][1,2,5]Methyl triazacyclononan-2-yl) acetate. MS (m/z) 599.3[ M+H ]] +
Step 3: synthesis of (1S, 2S, 5S) -8- (benzyloxy) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10- Formamide
((1S, 2S, 5S) -8- (benzyloxy) -5-methyl-7, 9-dioxo-10- ((2, 4, 6-trifluorobenzyl) carbamoyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Methyl triazacyclononan-2-yl) acetate (33 mg,0.06 mmol) was dissolved in MeOH (1 mL) and K2CO3 (24 mg,0.18 mmol) was added. The reaction was stirred at room temperature for 15 minutes. EtOAc was added to the reaction crude. Transferred to a separate funnel and water was added to wash the organic layer twice. The organic layer was concentrated and purified by silica column eluting with EtOAc/hexanes (60% -100%) to give (1 s,2s,5 s) -8- (benzyloxy) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluoro)Benzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 557.3[ M+H ]] +
Step 4: synthesis of (1S, 2S, 5S) -8- (benzyloxy) -2-formyl-5-methyl-7, 9-dioxo-N- (2, 4,6- Trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-A Amides and their use
(1S, 2S, 5S) -8- (benzyloxy) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10-carboxamide (22 mg,0.039 mmol) was dissolved in DCM (2 mL) and dess-Martin periodate (50 mg,0.12 mmol) was added. The reaction was stirred at room temperature and 50mg of dess-martin periodate was added later. After 4 hours the reaction was complete. Adding 10% Na 2 S 2 O 3 The solution was quenched. The crude product was extracted with DCM. The organic layer was concentrated and used directly in the next step. MS (m/z) 555.3[ M+H ]] +
Step 5: synthesis of (1S, 2S, 5S) -8- (benzyloxy) -2- (difluoromethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide
Crude (1S, 2S, 5S) -8- (benzyloxy) -2-formyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide was dissolved in DCM (1.5 mL) and Deoxofluor (50% in toluene, 2.7M,0.043 mL) was added. The reaction was stirred at room temperature and 0.043mL of deoxyfluor (50% toluene solution, 2.7M) was added. The reaction was stirred at room temperature overnight. Addition of saturated NaHCO 3 The solution was used to quench the reaction. Extraction using DCM. The organic layer was concentrated and purified by preparative HPLC eluting with 10% to 60% acetonitrile (0.1% tfa) in water (0.1% tfa) to give (1 s,2s,5 s) -8- (benzyloxy) -2- (difluoromethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1, 2-b) ][1,2,5]Triaza-sCyclononane-10-carboxamide. MS (m/z) 577.3[ M+H ]] +
Step 6: synthesis of (1S, 2S, 5S) -2- (difluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4,6- Trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-A Amides and their use
(1S, 2S, 5S) -8- (benzyloxy) -2- (difluoromethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (15 mg,0.026 mmol) was dissolved in toluene (0.5 mL) and TFA (0.5 mL) was added. The reaction was stirred at room temperature for 4 hours. Concentration of the reaction and purification via preparative HPLC eluting with 10% -60% acetonitrile (0.1% TFA) in water (0.1% TFA) afforded (1S, 2S, 5S) -2- (difluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1, 2-b)][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 487.2[ M+H ]] + .1H NMR (400 MHz, acetonitrile-d 3) δ10.26 (s, 1H), 8.44 (s, 1H), 6.87 (t, J=8.5 Hz, 2H), 6.11 (td, J=55.1, 3.5Hz, 1H), 4.72-4.54 (m, 5H), 3.55 (td, J=14.9, 4.1Hz, 1H), 2.23-2.01 (m, 2H), 1.85 (ddd, J=13.9, 10.5,7.7Hz, 1H), 1.79-1.59 (m, 1H), 1.26 (d, J=6.9 Hz, 3H).
Example 79: preparation of (1S, 2S, 5S) -8-hydroxy-2- (methoxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-A Amide (C79)
Step 1: synthesis of (1S, 2S, 5S) -8- (benzyloxy) -2- (methoxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide
(1S, 2S, 5S) -8- (benzyloxy) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 76][1,2,5]Triazacyclononane-10-carboxamide (46 mg,0.083 mmol) was dissolved in anhydrous DMF (2 mL), cooled to 0deg.C, and NaH (60%, 6mg,0.014 mL) was added. The reaction was kept at 0℃for 10 minutes. Diluted MeI (1.2 eq) was added. The reaction was kept at 0 ℃ for 10 minutes, then warmed to room temperature, and kept for 30 minutes. A drop of water was added to quench the reaction. The crude reaction was taken up in EtOAc/saturated NaHCO 3 And (5) extracting the solution. The organic layer was concentrated and purified by silica column eluting with EtOAc/hexanes (60% -100%) to give (1 s,2s,5 s) -8- (benzyloxy) -2- (methoxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] ][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 569.3[ M+H ]] +
Step 2: synthesis of (1S, 2S, 5S) -8-hydroxy-2- (methoxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxylic acid Amines
(1S, 2S, 5S) -8- (benzyloxy) -2- (methoxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (2 mg, 0.04 mmol) was dissolved in toluene (0.5 mL) and TFA (0.5 mL) was added. The reaction was stirred at room temperature for 4 hours. Concentration of the reaction and purification via preparative HPLC eluting with 10% -60% acetonitrile (0.1% TFA) in water (0.1% TFA) afforded (1S, 2S, 5S) -8-hydroxy-2- (methoxymethyl) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1, 2-b)][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 479.2[ M+H ]] + .1H NMR (400 MHz, acetonitrile-d 3) delta 8.59 (s, 1H), 6.84 (s, 1H), 6.70 (s, 1H), 5.77 (d, J=12.5 Hz, 1H), 5.41 (d, J=41.9 Hz, 2H), 4.96 (m, 1H), 4.67 (d, J=28.6 Hz, 3H), 3.95 (m, 1H), 3.66-3.53 (m, 2H), 3.36 (s, 3H), 1.47-1.15 (m, 3H).
Example 80: preparation of (1R, 2R, 5S) -2 - (fluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-) Trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C80)
(1R, 2R, 5S) -2- (fluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide to (1S, 2S, 5S) -2- (fluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] in example 76][1,2,5]Triazacyclononane-10-carboxamide is prepared in a similar manner except that the (S, S) -DACH naphthyl Trost ligand is used instead of the (R, R) -DACH naphthyl Trost ligand in step 3. MS (m/z) 467.2[ M+H] + .1H NMR (400 MHz, acetonitrile-d 3) delta 10.25 (s, 1H), 8.45 (d, J=5.4 Hz, 1H), 6.99-6.80 (m, 2H), 6.03-5.93 (m, 1H), 5.41 (ddd, J=12.0, 3.6,2.5Hz, 1H), 4.94-4.78 (m, 1H), 4.78-4.57 (m, 5H), 4.37-4.15 (m, 2H), 1.88-1.71 (m, 3H).
Example 81: preparation of (1S, 2S, 5S) -N- (2, 4-difluorobenzyl) -2- (difluoromethyl) -8-hydroxy-5-methyl-) 7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10- Formamide (C81)
Step 1: synthesis of ((1S, 2S, 5S) -8- (benzyloxy) -10- ((2, 4-difluorobenzyl) carbamoyl) -5-methyl) 1, 2-b-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2 ]][1,2,5]Triazacyclononane (T-N-phenylmethane) 2-yl) acetic acid methyl ester
((1S, 2S, 5S) -8- (benzyl) prepared in a similar manner to example 76Oxy) -10- ((2, 4-difluorobenzyl) carbamoyl) -5-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Methyl triazacyclononan-2-yl) acetate (170 mg, 0.284 mmol) was dissolved in 5ml ethanol and 5ml ethyl acetate and sparged under an argon atmosphere. Platinum (IV) oxide (34 mg,0.15 mmol) was added and the mixture was sparged under a hydrogen atmosphere (1 atm, balloon). The mixture was stirred for 20 minutes. After spraying with argon, passingThe pad was filtered and washed with absolute ethanol. The filtrate was concentrated to dryness and used directly in the next step. MS (m/z): 581.300[ M+H ]] +
Step 2: synthesis of (1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-A Amides and their use
Methyl ((1S, 2S, 5S) -8- (benzyloxy) -10- ((2, 4-difluorobenzyl) carbamoyl) -5-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononan-2-yl) acetate (170 mg,0.293 mmol) was dissolved in 10ml methanol. Potassium carbonate (80.9 mg,0.589 mmol) was added thereto. The reaction mixture was stirred at room temperature for 10 minutes and partitioned between ethyl acetate and water. The organic layer was separated, washed with brine, dried over magnesium sulfate and concentrated to dryness. The residue was dried under high vacuum to give the title product. MS (m/z): 539.300[ M+H ] +.
Step 3: synthesis of (1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-formyl-5-methyl-7, 9- Dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxylic acid Amines
To (1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (68 mg,0.126 mmol) in anhydrous DCM (7 ml) was addedGadess-Martin periodate (80.3 mg,0.189 mmol) and the mixture was stirred at room temperature for 30 min. DCM was added and the organic phase was washed twice with 10% sodium thiosulfate solution and once with brine. The organic phase was dried, evaporated to dryness and used directly in the next step. MS (m/z): 537.288[ M+H ]] +
Step 4-5: synthesizing (1S, 2S, 5S) -N- (2, 4-difluorobenzyl) -2- (difluoromethyl) -8-hydroxy-5-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-A Amides and their use
(1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2-formyl-5-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] in DCM (2 mL)][1,2,5]Triazacyclononane-10-carboxamide (30 mg,0.0559 mmol) was cooled to 0deg.C under argon. Deoxyfluor (50% toluene solution, 0.103ml,0.280 mmol) was added thereto under argon. The resulting mixture was stirred at 0 ℃ for 1 hour. The reaction mixture was diluted with DCM, cooled in an ice/water bath and quenched by dropwise addition of saturated aqueous NaHCO 3. The resulting mixture was stirred for 20 minutes. More saturated aqueous NaHCO3 was added until no more bubbling occurred. The organic layer was separated, dried over Na2SO4, and the solvent was removed under reduced pressure. The residue was purified by RP-HPLC eluting with ACN/water (containing 0.1% tfa) to give (1 s,2s,5 s) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (difluoromethyl) -5-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ][1,2,5]Triazacyclononane-10-carboxamide. The residue was dissolved in 1ml toluene and 1ml TFA and stirred at room temperature for one hour. The solvent was removed and the residue was purified by RP-HPLC eluting with ACN/water (containing 0.1% tfa) to give the title product. MS (m/z): 469.200[ M+H ]] +1 H NMR (400 MHz, acetonitrile-d 3) delta 10.26 (s, 1H), 8.45 (s, 1H), 7.49-7.38 (m, 1H), 7.03-6.92 (m, 2H), 6.11 (td, J=55.1, 3.6Hz, 1H), 4.72-4.57 (m, 5H), 3.62-3.49 (m, 1H), 2.18-1.99 (m, 2H), 1.88-1.72 (m, 2H), 1.27 (d, J=6.9 Hz, 3H).
Example 82: preparation of (1S, 2S, 5S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2- (methoxymethyl) -5-methyl 1, 2-b-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido][1,2,5]Triazacyclononane-10-A Amide (C82)
Step 1: synthesis of (1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide was prepared in a similar manner to example 81 step 2 except that ((1S, 2S, 5S) -8- (benzyloxy) -10- ((2, 4-difluorobenzyl) carbamoyl) -5-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononan-2-yl) acetic acid methyl ester was used in place of ((1S, 2S, 5S) -8- (benzyloxy) -10- ((2, 4-difluorobenzyl) carbamoyl) -5-methyl-7, 9-dioxo-2, 3, 7, 9-tetrahydro-1, 6-methano [1,2-b ] [1,2,5] triazacyclononan-2-yl) acetic acid methyl ester. MS (m/z): 537.300[ M+H ] +.
Step 2: synthesis of (1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (methoxymethyl) -5-methyl 1, 2-b-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido][1,2,5]Triazacyclononane-10-A Amides and their use
To a glass vial containing (1 s,2s,5 s) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (hydroxymethyl) -5-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (25 mg,0.0466 mmol) was added DMF (2.5 mL) under argon and cooled to 0 ℃. Sodium hydride (60% dispersion in mineral oil, 2.42mg,0.0606 mmol) was added and stirred at 0 ℃ for 20 min. Methyl iodide (0.0029 ml,0.0466 mmol) was added and stirred for 20 minutes. The reaction was quenched with saturated ammonium chloride solution and extracted into ethyl acetate. The organic phase was washed with water and then brine. The combined aqueous phases were back-extracted with more ethyl acetate. The combined organic phases were dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel to give the title product. MS (m/z): 551.300[ M+H ] +.
Step 3: synthesizing (1S, 2S, 5S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2- (methoxymethyl) -5-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (methoxymethyl) -5-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (12 mg,0.0218 mmol) was dissolved in 1ml toluene and 1ml TFA and stirred at room temperature for three hours. The solvent was removed and the residue was purified by RP-HPLC to give the title product. MS (m/z) 461.200[ M+H ] +.1H NMR (400 MHz, acetonitrile-d 3) delta 10.23 (s, 1H), 8.55 (s, 1H), 7.54-7.38 (m, 1H), 6.98 (ddt, J=13.0, 8.4,3.1Hz, 2H), 5.75 (dt, J=11.7, 2.7Hz, 1H), 5.38 (dt, J=12.1, 2.8Hz, 2H), 4.95 (d, J=14.4 Hz, 1H), 4.65-4.57 (m, 3H), 3.91 (dt, J=7.8, 4.3Hz, 1H), 3.63-3.48 (m, 2H), 3.36 (s, 3H), 1.35 (d, J=7.4 Hz, 3H).
Example 83: preparation of (1S, 2S, 5S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2- (methoxymethyl) -5-methyl 1, 2-b-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2 ]][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide (C83)
(1S, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2- (methoxymethyl) -5-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 82 ][1,2,5]Triazacyclononane-10-carboxamide (9 mg,0.0163 mmol) was dissolved in 3ml ethanol and 3ml ethyl acetate and under argon atmosphereAnd (5) lower spraying. Palladium on carbon (10 wt%, 2 mg) was added and the mixture was sparged under a hydrogen atmosphere (1 atm, balloon). The mixture was vigorously stirred for one hour and then sprayed under an argon atmosphere. By passing throughThe pad was filtered and washed with absolute ethanol. The filtrate was concentrated to dryness. The residue was purified by RP-HPLC to give the title product. MS (m/z) 463.200[ M+H ]]+.1H NMR (400 MHz, acetonitrile-d 3) delta 10.33 (s, 1H), 8.41 (s, 1H), 7.44 (td, J=8.8, 6.7Hz, 1H), 7.03-6.91 (m, 2H), 4.66 (d, J=14.9 Hz, 1H), 4.64-4.50 (m, 4H), 3.65 (dd, J=9.9, 7.0Hz, 1H), 3.51-3.34 (m, 2H), 3.38 (s, 3H), 2.03 (dd, J=14.7, 7.4Hz, 1H), 1.90-1.64 (m, 2H), 1.56 (ddd, J=15.2, 10.9,3.3Hz, 1H), 1.26 (d, J=6.8 Hz, 3H).
Example 84 and example 85: preparation of (1 'S,3S,5' S) -8 '-hydroxy-5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) -4,5,7',9' -tetrahydro-2H, 5 'H-spiro [ furan-3, 2' - [1,6 ]]Methanopyrido [1,2- ] b][1,2,5]Triazacyclononane]-10' -carboxamide and (1 ' S,3R,5' S) -8' -hydroxy-5 ' -methyl-7 ',9' -dioxo- N- (2, 4, 6-trifluorobenzyl) -4,5,7',9' -tetrahydro-2H, 5 'H-spiro [ furan-3, 2' - [1,6 ] ]A bridged methylene pyrido group [1 ], 2-b][1,2,5]triazacyclononane]-10' -carboxamide (C84 and C85)
Step 1: preparation of di-tert-butyl 1- (3-formyltetrahydrofuran-3-yl) hydrazine-1, 2-dicarboxylate
A mixture of tetrahydrofuran-3-carbaldehyde (2.04 g,0.02 mol), di-tert-butyl azodicarboxylate (3.13 g,0.014 mol) and L (-) -proline (0.6276 g,0.54 mmol) in DCE (50 mL) was stirred at 65℃for 3 days. The reaction mixture was concentrated and the residue was purified by silica gel chromatography eluting with 0% to 60% hexanes/EtOAc to give the title compound.
Step 2: preparation of 1- (3-vinyl tetra)Di-tert-butyl hydrofuran-3-yl) hydrazine-1, 2-dicarboxylic acid
To a suspension of methyltriphenyl phosphonium iodide (4.77 g,11.8 mmol) in THF (50 mL) was added potassium tert-butoxide (1.32 g,11.8 mmol) at 0deg.C. The reaction mixture was stirred at 0 ℃ for 5 minutes. To the mixture was added a solution of di-tert-butyl 1- (3-formyltetrahydrofuran-3-yl) hydrazine-1, 2-dicarboxylate, tert-butyl N- (tert-butoxycarbonylamino) -N- (3-formyltetrahydrofuran-3-yl) carbamate (1.3 g,3.93 mmol) in THF (5 mL) at 0deg.C. The reaction mixture was then stirred at room temperature for 1 hour. The reaction was treated with NH at 0deg.C 4 Aqueous Cl quench, extract with EtOAc and get Na anhydrous 2 SO 4 And (5) drying. The combined organic layers were concentrated under reduced pressure to give the crude product, which was then purified by silica column chromatography using hexane/acetate (0% -60%) as eluent to give the title compound.
Step 3: preparation of (3-vinyl tetrahydrofuran-3-yl) hydrazines
A reaction mixture of di-tert-butyl 1- (3-vinyltetrahydrofuran-3-yl) hydrazine-1, 2-dicarboxylate (82mg, 3.59 mmol) in DCM (3 mL) and TFA (3 mL) was stirred at room temperature for 1 hour. The reaction mixture was concentrated and used directly in the next step without purification.
Step 4: preparation of 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1- ((3-ethylene Methyl-3-yl-tetrahydrofuran-amino) -1, 4-dihydropyridine-2-carboxylate
To a solution of (3-vinyltetrahydrofuran-3-yl) hydrazine (460 mg,3.59 mmol) in MeOH (6 mL) and water (1 mL) was added 3-benzyloxy-4-oxo-5- [ (2, 4, 6-trifluorophenyl) methylcarbamoyl]Methyl pyran-2-carboxylate (1.6 g,3.59 mmol) and sodium bicarbonate (1.64 g,19.6 mmol). The reaction mixture was stirred at 60 ℃ overnight. The reaction mixture was cooled and the solvent was removed under vacuum. The residue was washed with water, extracted with EtOAc, the organic phase separated, and taken up over MgSO 4 Drying, filtration, concentration and purification by silica gel chromatography eluting with 0% to 100% hexanes/EtOAc afforded the title compound. MS (m/z) 558.04[ M+H ] ]+。
Step 5: preparation of 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1- ((3-ethylene Polytetrahydrofuran-3-yl) amino) -1, 4-dihydropyridine-2-carboxylic acid
A reaction mixture of 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1- ((3-vinyltetrahydrofuran-3-yl) amino) -1, 4-dihydropyridine-2-carboxylic acid methyl ester (760 mg,1.36 mmol) and lithium hydroxide monohydrate (284 mg,6.82 mmol) in THF (6 mL), meOH (6 mL) and water (2 mL) was stirred at 60℃for 4.5 h. The solvent was removed under vacuum. The residue was washed with 1N HCl and extracted with DCM. The organics were dried over MgSO 4 Drying, filtering and concentrating. The material was used in the next step without purification. MS (m/z) 544.11[ M+H ]]+。
Step 6: preparation of 3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) 1- ((3-vinyl tetrahydrofuran-3-yl) amino) -1, 4-dihydropyridine-2, 5-dicarboxamide
To a solution of 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1- ((3-vinyltetrahydrofuran-3-yl) amino) -1, 4-dihydropyridine-2-carboxylic acid (530 mg,0.975 mmol) in DMF (3 mL) was added HATU (1.0 g,2.53 mmol) and DIEA (786 mg,6.09 mmol) at 0 ℃. The reaction mixture was then stirred at room temperature for 1 hour. (S) -but-3-en-2-amine HCl salt (315 mg,2.92 mmol) was then added in one portion at room temperature. The reaction mixture was stirred at room temperature for 5 hours. The reaction mixture was treated with saturated NH 4 Washed with Cl and extracted with EtOAc. The organic phase was separated over MgSO 4 Drying, filtering and concentrating. The residue was purified by silica gel chromatography eluting with 0% to 80% hexanes/EtOAc to give the title compound. MS (m/z) 597.10[ M+H ]]+。
Step 7: preparation of 3- ((S) -but-3-en-2-yl) -5-hydroxy-4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) scheme 1- (3-vinyl tetrahydrofuran-3-yl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
To a solution of 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -1- (3-vinyltetrahydrofuran-3-yl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (791 mg,1.33 mmol) in DCE (10 mL)/ACN (10 mL) was added paraformaldehyde (139 mg,4.64 mmol), acetic acid (557 mg,9.28 mmol) and TFA (7516 mg,6.63 mmol). The reaction mixture was then stirred at room temperature and then the temperature was increased to 82 ℃. The reaction mixture was stirred at 82 ℃ for one day. Further paraformaldehyde (99 mg,3.31 mmol), acetic acid (557 mg,9.28 mmol) and TFA (756 mg,6.63 mmol) were then added to the mixture. The mixture was heated at 82 ℃ for an additional day. The reaction was cooled. The reaction mixture was concentrated and the residue was purified by silica gel chromatography eluting with 0% to 100% hexanes/EtOAc to give the title compound. MS (m/z) 519.22[ M+H ] +.
Step 8: preparation of 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) 1- (3-vinyl tetrahydrofuran-3-yl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxylic acid Amines
To 3- ((S) -but-3-en-2-yl) -5-hydroxy-4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -1- (3-vinyltetrahydrofuran-3-yl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]To a solution of triazine-7-carboxamide (460 mg,1.33 mmol) in DMF (10 mL) was added potassium carbonate (920 mg,6.65 mmol) and benzyl bromide (683 mg,3.99 mmol). The reaction mixture was stirred at 65 ℃ for 3 hours. The reaction was cooled. The reaction mixture was washed with water and extracted with EtOAc. The organic phase was dried over MgSO 4 Drying, filtering and concentrating. The residue was purified by silica gel chromatography eluting with 0% to 100% hexanes/EtOAc to give the title compound. MS (m/z) 609.12[ M+H]+。
Step 9: preparation of (1 'S,3S,5' S) -8'- (benzyloxy) -5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluoro) Benzyl) -4,5,7',9' -tetrahydro-2 h,5 'h-spiro [ furan-3, 2' - [1,6 ]]Methanopyrido [1,2-b ] bridged][1,2,5]Three kinds of Azacyclononane]-10' -carboxamide (A) and (1 ' S,3R,5' S) -8' - (benzyloxy) -5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) -4,5,7',9' -tetrahydro-2H, 5 'H-spiro [ furan-3, 2' - [1,6 ] ]Methanopyrido [1,2- ] b][1,2,5]Triazacyclononane]-10' -carboxamide (B)
A solution of 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -1- (3-vinyltetrahydrofuran-3-yl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (370 mg,0.608 mmol) in DCE (10 mL) was sparged with argon for 5 minutes. To the mixture was then added dichloro [1, 3-bis (2, 4, 6-trimethylphenyl) -2-imidazolidine ] (3-phenyl-1H-inden-1-ylidene) (tricyclohexylphosphine) ruthenium (II) (86.5 mg,0.091 mmol) and the mixture was sparged under argon for 5 minutes. The mixture was stirred at 80 ℃ for one day. To the mixture was added more dichloro [1, 3-bis (2, 4, 6-trimethylphenyl) -2-imidazolidinylidene ] (3-phenyl-1H-inden-1-ylidene) (tricyclohexylphosphine) ruthenium (II) (86.5 mg,0.091 mmol), sparged with argon and the reaction mixture was stirred at 80 ℃ for one week. The reaction mixture was concentrated and purified by silica gel chromatography eluting with 0% to 100% hexanes/EtOAc. Two desired compounds are formed in the reaction. The main product can be isolated as a pure single diastereomer by silica gel chromatography using 0% -100% hexane/EtOAc. The pure diastereomeric form of the minor product was isolated by SFC chiral separation.
The major diastereoisomer: MS (m/z) 581.13[ M+H ] +.
Minor diastereoisomers: MS (m/z) 581.09[ M+H ] +.
Step 10: preparation of (1 'S,3S,5' S) -8 '-hydroxy-5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -4,5,7',9' -tetrahydro-2 h,5 'h-spiro [ furan-3, 2' - [1,6]]Methanopyrido [1,2-b ] bridged][1,2,5]Trinitrogen Heterocyclononane]-10' -carboxamide and (1's, 3r, 5's) -8' -hydroxy-5 ' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluoro) Benzyl) -4,5,7',9' -tetrahydro-2 h,5 'h-spiro [ furan-3, 2' - [1,6]]Methanopyrido [1,2-b ] bridged][1,2,5]Three kinds of Azacyclononane]-10' -carboxamide
To a solution of (1's, 3s, 5's) -8' - (benzyloxy) -5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) -4,5,7',9' -tetrahydro-2 h,5' h-spiro [ furan-3, 2' - [1,6] methanopyrido [1,2-b ] [1,2,5] triazacyclononane ] -10' -carboxamide (a) in DMF was added LiCl (10 eq). The reaction mixture was heated at 100 ℃ overnight. The reaction mixture was filtered and the solution purified by reverse phase HPLC eluting with 5% -100% acetonitrile/water. (1 ' S,3R,5' S) -8' -hydroxy-5 ' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) -4,5,7',9' -tetrahydro-2H, 5' H-spiro [ furan-3, 2' - [1,6] methanopyrido [1,2-B ] [1,2,5] triazacyclononane ] -10' -carboxamide was synthesized in a similar manner except (1 ' S,3R,5' S) -8' - (benzyloxy) -5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) -4,5,7',9' -tetrahydro-2H, 5' H-spiro [ furan-3, 2' - [1,6] methanopyrido [1,2-B ] [1,2,5] triazacyclononane ] -10' -carboxamide (B) was used instead of (1 ' S,5' S) -8' - (benzyloxy) -5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) -4, 5', 9' -tetrahydro-2H, 5' H-spiro [ furan-3, 2' - [1,6] methanopyrido [1,2-B ] [1, 5] triazacyclononane ] -10' -carboxamide.
The major diastereoisomer: MS (m/z) 491.19[ M+H] + 1H NMR (400 MHz, methanol-d 4) delta 8.53 (s, 1H), 6.92 (t, J=8.4 Hz, 2H), 5.68 (dd, J=12.3, 2.7Hz, 1H), 5.53 (dd, J=12.3, 2.2Hz, 1H), 5.50-5.38 (m, 1H), 5.19 (d, J=14.5 Hz, 1H), 4.77 (d, J=14.5 Hz, 1H), 4.68 (s, 2H), 4.09 (dd, J=8.7, 1.3Hz, 1H), 3.79 (pd, J=8.9, 6.0Hz, 2H), 3.67 (d, J=8.7 Hz, 1H), 1.94 (ddd, J=14.8, 9.3,5.8, 1H), 1.51 (dt, J=14.5, 7.7 Hz, 1H), 4.09 (dd, J=8.7, 1 Hz, 3Hz, 1H).
Minor diastereoisomers: MS (m/z) 491.16[ M+H] + 1H NMR (400 MHz, methanol-d 4) delta 8.52 (s, 1H), 6.91 (t, J=8.4 Hz, 2H), 5.80 (dd, J=12.5, 3.7Hz, 1H), 5.48-5.38 (m, 1H), 5.34 (dd, J=12.5, 1.7Hz, 1H), 5.13 (s, 1H), 4.86 (s, 1H), 4.67 (s, 2H), 4.31-4.21 (m, 1H), 4.11 (td, J=8.9, 2.6Hz, 1H), 3.49 (d, J=11.0 Hz, 1H), 3.32-3.26 (m, 1H), 2.58 (dd, J=13.2, 7.0Hz, 1H), 2.25 (dt, J=13.2, 9.3Hz, 1H), 1.43 (d, J=7.7 Hz, 1H).
Example 86: preparation of (1 'S,5' S) -8 '-hydroxy-5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -4,4', 5',7',9' -hexahydro-2H, 3 'H-spiro [ furan-3, 2' - [1,6 ]]Methanopyrido [1,2-b ] bridged][1,2, 5]Triazacyclononane]-10' -carboxamide (C86)
To (1 'S,5' S) -8'- (benzyloxy) -5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl-4, 5,7',9' -tetrahydro-2H, 5 'H-spiro [ furan-3, 2' - [1,6 ] ]Methanopyrido [1,2-b ] bridged][1,2,5]Triazacyclononane]To a solution of 10' -carboxamide (10 mg,0.017 mmol) (major diastereomer prepared according to step 9 of example 84 and example 85) in EtOH (1 mL) was added platinum dioxide (2 mg). The reaction mixture was taken up at room temperature at H 2 Stir under balloon overnight. The reaction mixture was filtered through celite, the filtrate was concentrated and the residue was purified by reverse phase HPLC eluting with 5% -100% acetonitrile/water to give the title compound. MS (m/z) 493.17[ M+H ]] + .1H NMR (400 MHz, methanol-d 4) delta 8.44 (s, 1H), 6.97-6.87 (m, 2H), 4.81 (d, J=14.9 Hz, 1H), 4.75-4.60 (m, 3H), 4.16 (q, J=7.7 Hz, 2H), 3.95 (td, J=8.9, 5.0Hz, 1H), 3.68 (d, J=9.4 Hz, 1H), 2.18 (dt, J=14.6, 6.5Hz, 1H), 1.92-1.56 (m, 6H), 1.30 (d, J=6.8 Hz, 3H).
Example 87: preparation of (1S, 5S) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-AAmide (C87)
Synthesis of (1S, 5S) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
(1S, 5S) -8- (benzyloxy) -2, 5-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 48 ][1,2,5]Triazacyclononane-10-carboxamide (0.005 g, 0.09 mmol,1 eq.) was dissolved in methanol (1 mL) and platinum (IV) oxide (0.2 mg,0.001mmol,0.1 eq.) was added. The vials are sealedSeal, evacuate, and then backfill with hydrogen (repeat 2 times). The reaction mixture was sparged with hydrogen for 5 minutes and stirred under 1atm hydrogen for 4 hours. The reaction mixture was filtered and concentrated to give a crude residue, which was dissolved in MeCN, filtered and purified by preparative HPLC (column, gemini 10 μc18 a, axi/;250×21.2 mm), eluting with 5% -100% acetonitrile (0.1% tfa) in water (0.1% tfa) over 20 minutes. The combined fractions were lyophilized to give (1S, 5S) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z) 465.21[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δ10.34(t,J=6.0Hz,1H),8.28(s,1H),7.21(t,J=8.7Hz,2H),4.79(d,J=14.8Hz,1H),4.65(d,J=14.8Hz,1H),4.56(d,J=5.7Hz,2H),4.53–4.44(m,1H),1.96–1.84(m,1H),1.71–1.58(m,1H),1.49(dd,J=15.4,6.7Hz,1H),1.32(s,3H),1.26–1.22(m,1H),1.20(d,J=6.8Hz,3H),0.92(s,3H)。 19 F NMR(376MHz,DMSO-d 6 )δ-109.16–-109.37(m),-112.53(t,J=7.3Hz)。
Example 88 and example 89: preparation of (1 aS,2R,3S,11 aR) -8-hydroxy-2, 11-dimethyl-7, 9-di- oxo-N- (2, 4, 6-trifluorobenzyl) -1a,2,7,9,11 a-hexahydro-1H-3, 10-methanocyclopropane [ g ]]Pyrido-s [1,2-b][1,2,5]Triazacyclononane-6-carboxamide and (1 aS,2R,3R,11S,11 aR) -8-hydroxy-2, 11-dimethyl-) 7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -1a,2,7,9,11 a-hexahydro-1H-3, 10-methanocyclopropane [ g ] ] Pyrido [1,2-b][1,2,5]Triazacyclononane-6-carboxamide (C88 and C89)
Step 1: preparation of (S) -but-3-en-2-yl-phenyl ((S) -but-3-en-2-yl) phosphoramidate
(S) -but-3-en-2-amine hydrochloride (1 equivalent, 23.2mmol,2.5 g) and phenyl dichlorophosphate (1 equivalent, 23.2mmol,3.47 mL) in 40mL DCM was cooled to-78deg.C under argon, treated dropwise with triethylamine (2 eq., 46.5mmol,6.5 mL) and allowed to slowly warm to room temperature overnight. The reaction mixture was then cooled again to-78 ℃, treated with (S) -but-3-en-2-ol (1.5 eq, 34.9mmol,2.5 g), followed by dropwise addition of NMI (2 eq, 46.5mmol,3.7 mL) in 10mL DCM and allowed to slowly warm to room temperature overnight. The reaction was quenched with water and extracted into EtOAc (3×). The combined organic layers were dried over sodium sulfate, filtered and concentrated. The crude mixture was purified by silica gel chromatography (0% -100% etoac in hexanes) to give the desired product as a-1:1 mixture of phosphorus-containing diastereomers. MS (m/z) 281.9[ M+H ]]+。 1 H NMR (400 MHz, chloroform-d) delta 7.36-7.18 (m, 4H), 7.17-7.09 (m, 1H), 5.96-5.74 (m, 2H), 5.37-4.92 (m, 4H), 4.01-3.81 (m, 1H), 2.75-2.60 (m, 1H), 1.46 (d, j=6.4 hz, 1.5H), 1.38 (d, j=6.4 hz, 1.5H), 1.25 (d, j=6.9 hz, 1.5H), 1.23 (d, j=6.8 hz, 1.5H). 31 P NMR (162 MHz, chloroform-d) delta 2.63-1.96 (m).
Step 2: preparation of (4S, 7S) -4, 7-dimethyl-2-phenoxy-3, 4, 7-dihydro-1, 3, 2-phosphacycle heptatriene 2- Oxide compound
A solution of (S) -but-3-en-2-yl phenyl ((S) -but-3-en-2-yl) phosphoramidate (11.7 mmol,3.29 g) in 1,2-DCE (470 mL) was sparged with argon for 20 minutes, treated with a generation 2 Hoveyda-Grubbs catalyst (0.05 eq, 366 mg) for a further 5 minutes, and then stirred at room temperature. Another portion of catalyst was added at about 24 hours and 48 hours in the same manner as described above. After 72 hours, the reaction mixture was adsorbed onto silica gel and purified by silica gel chromatography (0% -100% etoac in hexanes). Diastereomers elute in different bands, but they are combined and concentrated to give the desired product as a-1:1 mixture of phosphorus-containing diastereomers. Diastereomer 1 (LCMS rt=1.14 min): MS (m/z) 254.19[ M+H ]]+. Diastereomer 2 (LCMS rt=1.20 min): MS (m/z) 254.18[ M+H ]]+。 1 H NMR (400 MHz, chloroform-d) delta 7.37-7.11 (m, 5H), 5.55-5.35 (m, 2.5H), 5.24-5.13 (m, 0.5H), 4.24 (H, j=7.2 hz, 0.5H), 4.13-4.01 (m, 0.5H), 3.30 (dd, j=11.8, 7.0hz, 0.5H), 3.11 (t, j=6.7Hz,0.5H),1.45(dt,J=6.9,1.6Hz,3H),1.33(dd,J=7.1,2.2Hz,1.5H),1.25(dd,J=7.1,2.4Hz,1.5H)。 31 P NMR (162 MHz, chloroform-d) delta 6.08(s), 4.93 (d, j=8.8 Hz).
Step 3: preparation of tert-butyl ((2S, 5S, Z) -5-hydroxyhex-3-en-2-yl) carbamate
A solution of (4S, 7S) -4, 7-dimethyl-2-phenoxy-3, 4, 7-dihydro-1, 3, 2-phosphacycle hept-trien 2-oxide (6.32 mmol,1.6 g) in 60mL THF was cooled to-78℃under argon. Lithium aluminum hydride solution (2M THF,3.75 eq., 23.7mmol,11 mL) was added dropwise, and the reaction mixture was then allowed to slowly warm to room temperature overnight. The reaction was cooled to 0deg.C, then carefully quenched with 1mL of water, 1mL of 10% aqueous NaOH, and 1.5mL of water, warmed to room temperature, treated with magnesium sulfate, and passed through celite with additional CH 2 Cl 2 Together with filtration. The filtrate was concentrated to give 2s,5s, z) -5-aminohex-3-en-2-ol as a clear oil. 1 H NMR (400 MHz, chloroform-d) delta 5.48-5.31 (m, 2H), 4.66-4.57 (m, 1H), 3.91 (p, j=6.7 hz, 1H), 1.28 (d, j=6.5 hz, 3H), 1.22 (d, j=6.5 hz, 3H).
The crude reaction mixture containing (2 s,5s, z) -5-aminohex-3-en-2-ol was dissolved in 65mL DCM and cooled to 0 ℃ under argon, then treated with triethylamine (2 eq, 12.6mmol,1.76 mL) and di-tert-butyl dicarbonate (1.5 eq, 9.48mmol,2.07 g) and allowed to slowly warm to room temperature overnight. The reaction mixture was concentrated, then redissolved in EtOAc and washed with half-saturated aqueous sodium bicarbonate and brine, dried over sodium sulfate, filtered and concentrated. The crude residue was purified by silica gel chromatography (0% -100% etoac in hexanes) to afford the desired product as a white solid. MS (m/z) 215.76[ M+H ] ]+。 1 H NMR (400 MHz, chloroform-d) delta 5.46 (ddd, j=11.1, 7.3,1.0hz, 1H), 5.20 (t, j=10.3 hz, 1H), 4.83-4.61 (m, 2H), 4.45 (bs, 1H), 1.43 (s, 9H), 1.28 (d, j=6.4 hz, 3H), 1.20 (d, j=6.7 hz, 3H).
Step 4: preparation of t-butyl ((S) -1- ((1R, 2S) -2- ((S) -1-hydroxyethyl) cyclopropyl) ethyl) carbamate Esters
Diethyl zinc (1M hexane solution, 5 eq, 9.41mmol,9.4mL) On CH 2 Cl 2 The solution in (4 mL) was cooled to 0deg.C under argon, treated dropwise with trifluoroacetic acid (4.8 eq, 9.0mmol,690 μL) and stirred for 15 min. Diiodomethane (5 eq, 9.41mmol,760 μl) was added dropwise and the reaction mixture stirred for an additional 20 minutes. Tert-butyl ((2S, 5S, Z) -5-hydroxyhex-3-en-2-yl) carbamate (1 eq, 1.88mmol,405 mg) in CH 2 Cl 2 (2 mL) and the reaction mixture was allowed to slowly warm to room temperature overnight, then quenched with saturated ammonium chloride and extracted into EtOAc (3X). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The crude residue was purified by silica gel chromatography (0% -100% etoac in hexanes) to give the title compound. MS (m/z) 229.73[ M+H ]]+。 1 H NMR (400 MHz, chloroform-d) delta 4.57 (s, 1H), 3.80-3.66 (m, 1H), 3.50-3.33 (m, 1H), 1.43 (s, 9H), 1.30 (d, j=6.2 hz, 3H), 1.25 (d, j=6.3 hz, 3H), 0.99-0.79 (m, 2H), 0.74 (td, j=8.5, 4.5hz, 1H), 0.34 (d, j=5.7 hz, 1H).
Step 5: preparation of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) ((R) -1- ((1S, 2R) -2- ((S) -1- ((tert-butyl) Oxy carbonyl) amino) ethyl) cyclopropyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid methyl ester
A solution of ((S) -1- ((1R, 2S) -2- ((S) -1-hydroxyethyl) cyclopropyl) ethyl) carbamate (1.2 eq, 0.427mmol,98 mg), methyl 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylate (1 eq, 0.356mmol,200 mg) and triphenylphosphine (1.2 eq, 0.427mmol,106 mg) in THF (3.5 mL) was cooled under argon to 0deg.C and then treated dropwise with diisopropyl azodicarboxylate (1.2 eq, 0.427mmol, 85. Mu.L) and allowed to warm slowly to room temperature overnight. The reaction mixture was then treated with another portion of triphenylphosphine and diisopropyl azodicarboxylate at 0 ℃ and warmed again to room temperature overnight, then concentrated and purified by silica gel chromatography (0% -100% etoac in hexanes) followed by reversed phase C18 chromatography (0% -100% mecn in water). Clear to be combinedThe clean fraction was concentrated and then redissolved in CH 2 Cl 2 Dried over sodium sulfate, filtered and concentrated to give the title product. MS (m/z) 773.01[ M+H ]]+。
Step 6: preparation of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) ((R) -1- ((1S, 2R) -2- ((S) -1- ((tert-butyl) Oxy carbonyl) amino) ethyl) cyclopropyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid
A solution of 3- (benzyloxy) -1- ((tert-butoxycarbonyl) ((R) -1- ((1R, 2S) -2- ((S) -1- ((tert-butoxycarbonyl) amino) ethyl) cyclopropyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid methyl ester (0.113 mmol,87 mg) in 3/2/1 THF/MeOH/water (4 mL) was treated with lithium hydroxide (4 eq. 0.45mmol,19 mg) and stirred at room temperature overnight. The reaction mixture was treated with another portion of lithium hydroxide and further stirred at room temperature, then carefully acidified to pH-3 with 1N HCl and extracted into EtOAc (3×). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated to give the title compound which was carried out in step 7 without further purification. MS (m/z) 759.02[ M+H ] +.
Step 7: preparation of (1 aS,2R,11S,11 aR) -8- (benzyloxy) -2, 11-dimethyl-7, 9-dioxo-N- (2, 4), 6-trifluorobenzyl) -1a,2,3,7,9,10,11 a-octahydro-1H-cyclopropaneo [ g ]]Pyrido [1,2-b][1,2,5]Trinitrogen Heterocyclononane-6-carboxamide
The crude reaction mixture containing 3- (benzyloxy) -1- ((tert-butoxycarbonyl) ((R) -1- ((1R, 2S) -2- ((S) -1- ((tert-butoxycarbonyl) amino) ethyl) cyclopropyl) ethyl) amino) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid from step 6 was dissolved in CH 2 Cl 2 (200. Mu.L) and 4N HCl/dioxane (70. Mu.L) were stirred at room temperature for 3 hours, then concentrated and redissolved in CH 2 Cl 2 And concentrated again (3×) to give 1- (((R) -1- ((1R, 2S) -2- ((S) -1-aminoethyl) cyclopropyl) ethyl) amino) -3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl)Base) carbamoyl) -1, 4-dihydropyridine-2-carboxylic acid. MS (m/z) 559.28[ M+H ]]+. Dissolving the crude residue in CH 2 Cl 2 (1 mL) was treated with N, N-diisopropylethylamine (5 eq, 0.079mmol, 14. Mu.L) and 2- (7-aza-1H-benzotriazol-1-yl) -1, 3-tetramethyluronium hexafluorophosphate (HATU, 1.5 eq, 0.024mmol,9.0 mg) and then stirred at room temperature overnight. The reaction mixture was diluted with EtOAc, washed with saturated aqueous sodium bicarbonate solution, and then further extracted with EtOAc (2×). The combined organic layers were dried over sodium sulfate, filtered and concentrated. The crude residue was purified by silica gel chromatography (0% -100% etoac in hexanes) to give the title compound. MS (m/z) 541.10[ M+H ] ]+。
Step 8: preparation of (1 aS,2R,3S,11 aR) -8- (benzyloxy) -2, 11-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -1a,2,7,9,11 a-hexahydro-1H-3, 10-methanocyclopropane [ g ]]Pyrido [1,2-b] [1,2,5]Triazacyclononane-6-carboxamide and (1 aS,2R,3R,11S,11 aR) -8- (benzyloxy) -2, 11-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -1a,2,7,9,11 a-hexahydro-1H-3, 10-methanocyclopropane [ g ]]Piirae-type pyridine Pyrido [1,2-b][1,2,5]Triazacyclononane-6-carboxamide
A solution of (1 aS,2R,11S,11 aR) -8- (benzyloxy) -2, 11-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -1a,2,3,7,9,10,11 a-octahydro-1H-cyclopropaneo [ g ] pyrido [1,2-b ] [1,2,5] triazacyclononane-6-carboxamide (0.044 mmol,23.7 mg) in 1/1MeCN/1,2-DCE (0.8 mL) was treated with paraformaldehyde (3 eq, 0.13mmol,12 mg), acetic acid (7 eq, 0.31mmol, 18. Mu.L) and trifluoroacetic acid (5 eq, 0.22mmol, 17. Mu.L) and then heated to 85℃for 90 minutes. The reaction mixture was concentrated, then redissolved in EtOAc and washed with saturated aqueous sodium bicarbonate solution and the aqueous layer was further extracted with EtOAc (2×). The combined organic layers were dried over sodium sulfate, filtered and concentrated. The crude residue was purified by silica gel chromatography (0% -100% EtOAc in hexanes) followed by preparative TLC (EtOAc) to give the title compound as isolated diastereomers. Minor diastereoisomers (LCMS rt=1.51 min): MS (m/z) 553.07[ M+H ] +. Major diastereomer (LCMS rt=1.55 min): MS (m/z) 553.09[ M+H ] +.
Step 9: preparation of (1 aS,2R,3S,11 aR) -8-hydroxy-2, 11-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -1a,2,7,9,11 a-hexahydro-1H-3, 10-methanocyclopropaneo [ g ]]Pyrido [1,2-b][1, 2,5]Triazacyclononane-6-carboxamide (C88)
(1 aS,2R,3S,11 aR) -8- (benzyloxy) -2, 11-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -1a,2,7,9,11 a-hexahydro-1H-3, 10-methanocyclopropa [ g ]]Pyrido [1,2-b][1,2,5]A solution of triazacyclononane-6-carboxamide (0.0081 mmol,4.5 mg) (minor diastereomer from step 8) in DMF (0.15 mL) was treated with lithium chloride (10 eq., 0.081mmol,3.5 mg) and heated to 100deg.C for 6 hours. The reaction mixture was cooled to room temperature, diluted with MeOH/MeCN/TFA, filtered, purified by preparative HPLC (10% -100% MeCN in water, 0.1% TFA) and lyophilized to give the title compound. MS (m/z) 463.29[ M+H ]]+。 1 H NMR (400 MHz, chloroform-d) δ10.35 (t, j=4.9 hz, 1H), 8.48 (s, 1H), 6.73-6.60 (m, 2H), 5.13 (q, j=7.1 hz, 1H), 4.67 (s, 2H), 4.64 (d, j=8.9 hz, 1H), 4.49 (d, j=14.3 hz, 1H), 4.29 (p, j=6.9 hz, 1H), 1.50 (d, j=7.1 hz, 3H), 1.23-1.17 (m, 2H), 1.18 (d, j=7.0 hz, 3H), 1.12 (q, j=8.0 hz, 1H), 0.17 (q, j=6.9 hz, 1H).
Step 10: preparation of (1 aS,2R,3R,11S,11 aR) -8-hydroxy-2, 11-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -1a,2,7,9,11 a-hexahydro-1H-3, 10-methanocyclopropaneo [ g ]]Pyrido [1,2-b][1, 2,5]Triazacyclononane-6-carboxamide (C89)
(1 aS,2R,3R,11S,11 aR) -8- (benzyloxy) -2, 11-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -1a,2,7,9,11 a-hexahydro-1H-3, 10-methanocyclopropa [ g ]]Pyrido [1,2-b][1,2,5]A solution of triazacyclononane-6-carboxamide (0.01 mmol,5.5 mg) (major diastereomer from step 8) in EtOH (0.2 mL) was purged with argon, then treated with 10% palladium on carbon (2 mg), hydrogen balloon immobilized and purged with hydrogen (3X). 30 minutesAfter clock, the reaction mixture was filtered through celite, concentrated, purified by preparative HPLC (10% -100% mecn in water, 0.1% tfa) and lyophilized to give the title compound. MS (m/z) 463.21[ M+H ]]+。 1 H NMR (400 MHz, chloroform-d) δ10.40 (t, j=5.1 hz, 1H), 8.62 (s, 1H), 6.71-6.62 (m, 2H), 4.77-4.59 (m, 3H), 4.44 (d, j=15.0 hz, 1H), 4.44-4.34 (m, 1H), 3.01 (dq, j=10.0, 6.8hz, 1H), 1.94-1.86 (m, 1H), 1.85 (d, j=6.7 hz, 3H), 1.39 (d, j=7.5 hz, 3H), 1.12 (td, j=8.8, 5.4hz, 1H), 1.00 (q, j=5.7 hz, 1H), 0.91 (qd, j=8.7, 5.6hz, 1H).
Example 90: preparation of (1S, 2R) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo- 2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C90)
Step 1: preparation of (1S, 2R) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-trimethyl-7, 9-dioxo Substituted-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (A), (1R, 2S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6- Methanopyrido [1,2-b ] bridged][1,2,5]Triazacyclononane-10-carboxamide (B) and (1S, 2S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]] [1,2,5]Triazacyclononane-10-carboxamide (C)
5- (benzyloxy) -1- (but-3-en-2-yl) -N- (2, 4-difluorobenzyl) -3- (2-methylbut-3-en-2-yl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (400 mg,0.711 mmol) prepared at room temperature in analogy to example 29, except that 2-methylbut-3-en-2-amine was used instead of (S) -but-3-en-2-amine was dissolved in dichloromethane (60 mL). Argon was bubbled through the reaction solution for 20 minutes. HG-M720 catalyst (44.5 mg,0.071 mmol) was then added with stirring. Purging with argon was continued for 10 minutes. The reaction mixture was connected to a reflux condenser (evacuated, flushed with argon three times) and then heated under argon atmosphere with stirring for 24 hours. The resulting reaction mixture was then concentrated to dryness. The crude material was purified on a silica gel column with 0% -100% etoac/Hex to give three diastereomers. (1S, 2R) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (A) MS (m/z) 535.1[ M+H ] +. (1R, 2S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-B ] [1,2,5] triazacyclononane-10-carboxamide (B) MS (m/z) 535.0[ M+H ] +. (1S, 2S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (C) MS (m/z) 535.1[ M+H ] +.
Step 2: preparation of (1S, 2R) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (25 mg, 0.56 mmol) dissolved in CH 3 CN (3 mL) was added MgBr 2 (38 mg,0.206 mmol) and stirred at 50℃for 2 hours. The reaction mixture was quenched with water (1 mL) to form a clear solution, filtered and the residue was dissolved in MeOH, and taken up in 50% -100% CH 3 The CN aqueous solution was purified by reverse phase prep HPLC to give the desired product. MS (m/z): 445.1[ M+H ]]+.1H NMR (400 MHz, acetonitrile-d 3) delta 10.39 (s, 1H), 8.41 (s, 1H), 7.44 (td, J=9.2, 8.8,6.5Hz, 1H), 7.03-6.90 (m, 2H), 5.70-5.45 (m, 2H), 5.13 (d, J=14.5 Hz, 1H), 4.59 (dd, J=13.6, 5.4Hz, 4H), 1.83 (s, 3H), 1.45 (s, 3H), 1.03 (d, J=7.3 Hz, 3H).
Example 91 and example 92: preparation of (1R, 2S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl- 7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxylic acid Amine and (1S, 2S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6- Methanopyrido [1,2-b ] bridged][1,2,5]Triazacyclononane-10-carboxamide (C91 and C92)
(1R, 2S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 90][1,2,5]Triazacyclononane-10-carboxamide (B) and (1S, 2S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-B][1,2,5]A mixture of triazacyclononane-10-carboxamide (C) (25 mg,0.0.56 mmol) was dissolved in CH 3 CN (3 mL) and MgBr2 (38 mg,0.206 mmol) were added and stirred at 50℃for 2 hours. The reaction mixture was quenched with water (1 mL) to form a clear solution, filtered and the residue was dissolved in MeOH. The crude product is treated with 50% -100% CH 3 The aqueous solution of CN was purified by reverse phase preparative HPLC to give the desired product (1 r,2 s) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide and (1S, 2S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide.
(1 r,2 s) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (C91): MS (m/z): 445.1[ M+H ] +.1H NMR (400 MHz, acetonitrile-d 3) delta 10.35 (s, 1H), 8.37 (s, 1H), 7.44 (td, J=9.2, 8.8,6.5Hz, 1H), 6.97 (ddt, J=13.0, 8.5,3.0Hz, 2H), 5.67 (dd, J=11.2, 2.5Hz, 1H), 5.39 (dd, J=11.2, 4.1Hz, 1H), 5.11 (d, J=14.3 Hz, 1H), 4.60 (d, J=6.0 Hz, 2H), 4.51 (d, J=14.2 Hz, 1H), 3.93 (ddd, J=6.7, 4.1,2.6Hz, 1H), 1.76 (s, 3H), 1.42 (s, 3H), 1.33 (d, J=6.6 Hz, 3H).
(1S, 2S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (C92): MS (m/z): 445.1[ M+H ] +.1H NMR (400 MHz, acetonitrile-d 3) delta 10.35 (s, 1H), 8.37 (s, 1H), 7.52-7.32 (m, 1H), 6.97 (ddt, J=11.2, 8.6,3.0Hz, 2H), 5.67 (dd, J=11.2, 2.5Hz, 1H), 5.39 (dd, J=11.2, 4.1Hz, 1H), 5.11 (d, J=14.2 Hz, 1H), 4.60 (d, J=5.9 Hz, 2H), 4.51 (d, J=14.3 Hz, 1H), 3.93 (ddd, J=6.6, 4.1,2.5Hz, 1H), 1.76 (s, 3H), 1.33 (d, J=6.6 Hz, 3H).
Examples 93 and 94: preparation of (1S, 2R) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl- 7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10- Formamide and (1R, 2S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-2, 3,4,5,7,9- Hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (C93 and C94)
(1S, 2R) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 90][1,2,5]Triazacyclononane-10-carbonyl (A) and (1R, 2S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ][1,2,5]A mixture of triazacyclononane-10-carboxamide (15 mg,0.028 mmol) was dissolved in EtOH (3 mL) and 10% Pd-C (6 mg, 0.006mmol) was added. With H at room temperature 2 The balloon was subjected to hydrogenolysis for 7 hours. The reaction mixture was filtered through a celite pad. The filtrate was collected and concentrated to dryness. The residue was dissolved in MeOH and taken up in 50% -100% CH with 0.1% TFA 3 The aqueous solution of CN was purified by reverse phase preparative HPLC to give the desired product (1S, 2R) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-2, 3,4,5,79-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide and (1R, 2S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide.
(1S, 2R) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C93): MS (m/z): 447.2[ M+H ]]+。 1 H NMR (400 MHz, acetonitrile-d) 3 )δ10.45(s,1H),8.32(s,1H),7.44(d,J=7.6Hz,1H),6.97(t,J=9.9Hz,3H),5.00–4.43(m,4H),1.88–1.77(m,3H),1.68(s,2H),1.64–1.47(m,2H),1.39(s,3H),1.12(d,J=6.9Hz,3H)。
(1R, 2S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C94): MS (m/z): 447.1[ M+H ] ]+。 1 H NMR (400 MHz, acetonitrile-d) 3 )δ10.45(s,1H),8.32(s,1H),7.44(q,J=9.3,8.5Hz,1H),6.97(t,J=10.0Hz,2H),4.83(d,J=14.6Hz,1H),4.66(d,J=14.6Hz,1H),4.60(d,J=5.9Hz,2H),3.74–3.30(m,1H),1.74–1.52(m,4H),1.39(s,3H),1.30(s,3H),1.12(d,J=6.9Hz,3H)。
Example 95: preparation of (1S, 2S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo- 2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (C95)
(1S, 2S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -2, 5-trimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 90][1,2,5]Triazacyclononane-10-carboxamide (C) (20 mg,0.037 mmol) was dissolved in EtOH (3 mL) and 10% Pd-C (8 mg,0.0075 mmol) was added. Hydrogenolysis was performed with an H2 balloon at room temperature for 7 hours. The reaction mixture was filtered through a celite pad. The filtrate was collected and concentrated to dryness. The residue was dissolved in MeOH and taken up in 50% -100% CH with 0.1% TFA 3 The aqueous solution of CN was purified by reverse phase preparative HPLC to give the desired product (1S, 2S) -N- (2, 4-difluorobenzyl) -8-hydroxy-2, 5-trimethyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide. MS (m/z): 447.1[ M+H ]]+.1H NMR (400 MHz, acetonitrile-d 3) delta 8.57-8.18 (m, 1H), 7.45 (s, 1H), 7.01 (d, J=22.1 Hz, 2H), 6.77 (s, 1H), 5.78-5.26 (m, 1H), 5.19-4.14 (m, 4H), 3.97 (s, 1H), 1.87 (s, 4H), 1.82-1.67 (m, 3H), 1.46-1.31 (m, 3H), 1.30 (s, 3H).
Example 96: preparing (1R, 2S, 5S) -N- (2, 4-difluorobenzyl) -5- (fluoromethyl) -8-hydroxy-2-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-A Amide (C96)
Step 1: synthesis of 3- (benzyloxy) -1- (but-3-en-2-yl (tert-butoxycarbonyl) amino) -5- ((2, 4-difluoro) Benzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylic acid methyl ester
To a stirred mixture of methyl 3- (benzyloxy) -1- ((tert-butoxycarbonyl) amino) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylate (8.0 g,14.7 mmol) and but-3-en-2-ol (1.592 g,22.1 mmol) in THF (75 mL) was added triphenylphosphine (5.79 g,22.1 mmol) at room temperature. The resulting mixture was cooled to 0 ℃, diisopropyl azodicarboxylate (4.46 g,22.1 mmol) was added and the newly formed mixture was stirred at 0 ℃ for 10 min before it was removed from the cooling bath and stirred at room temperature for 1 hour. The reaction was then filtered through a pad of silica gel, rinsed with 60% etoac/hexanes, the filtrate was mixed with silica gel, concentrated and purified by normal phase chromatography. LCMS-esi+ (m/z): h+ calculated theory for C31H33F2N3O 7: 597.23, found: 597.87.
Step 2: synthesis of 3- (benzyloxy) -1- (butyl)-3-en-2-yl (t-butoxycarbonyl) amino) -5- ((2, 4-difluoro) Benzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylic acid
Methyl 3- (benzyloxy) -1- (but-3-en-2-yl (tert-butoxycarbonyl) amino) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylate (10.0 g,16.7 mmol) was dissolved in a mixture of MeOH (96 mL), THF (48 mL) and water (48 mL). Adding LiOH-H to the stirred mixture 2 O (4.21 g,100 mmol). The resulting mixture was heated to 60 ℃ for 6 hours. The reaction was then cooled to room temperature, concentrated, the residue diluted with EtOAc, acidified to pH-4 with 1N HCl, the layers separated, the organic layer washed with brine, dried over sodium sulfate, filtered and concentrated. LCMS-esi+ (m/z): h+ calculated theory for C30H31F2N3O 7: 583.21, found: 583.868.
step 3: synthesis of (3- (benzyloxy) -5- ((2, 4-difluorobenzyl) carbamoyl) -2- (((S) -1-hydroxybutan-e) 3-en-2-yl) carbamoyl) -4-oxopyridin-1 (4H) -yl) (but-3-en-2-yl) carbamic acid tert-butyl ester
To a mixture of 3- (benzyloxy) -1- (but-3-en-2-yl (tert-butoxycarbonyl) amino) -5- ((2, 4-difluorobenzyl) carbamoyl) -4-oxo-1, 4-dihydropyridine-2-carboxylic acid (1.5 g,2.57 mmol) and (2S) -2-aminobut-3-en-1-ol hydrochloride (381 mg,3.08 mmol) in DCM (12.0 mL) was added 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide HCl (386 mg,3.86 mmol) followed by 1-hydroxy-7-azabenzotriazole (525 mg,3.86 mmol) and DIEA (1.329 g,10.3 mmol) at room temperature. The resulting mixture was stirred at room temperature overnight. The reaction was then diluted with DCM, washed with 10% citric acid, brine, dried over sodium sulfate, filtered and concentrated. Purification by normal phase chromatography.
5 2 Step 4: synthesis of 3- (benzyloxy) -1- (but-3-en-2-ylamino) -N- (2, 4-difluorobenzyl) -N- ((S) -1- Hydroxybut-3-en-2-yl) -4-oxo-1, 4-dihydropyridine-2, 5-dicarboxamide
Tert-butyl (3- (benzyloxy) -5- ((2, 4-difluorobenzyl) carbamoyl) -2- (((S) -1-hydroxybut-3-en-2-yl) carbamoyl) -4-oxopyridin-1 (4H) -yl) (but-3-en-2-yl) carbamate (700 mg,1.07 mmol) was dissolved in DCM (10 mL) and treated with a solution of 4N HCl in 1, 4-dioxane (10 mL) for 90 min at room temperature. The reaction was concentrated, co-evaporated with ethyl acetate (×5), then co-evaporated with acetonitrile (×5). Directly used in the next step.
Step 5: synthesis of 5- (benzyloxy) -1- (but-3-en-2-yl) -N- (2, 4-difluorobenzyl) -3- ((S) -1-hydroxy But-3-en-2-yl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
3- (benzyloxy) -1- (but-3-en-2-ylamino) -N5- (2, 4-difluorobenzyl) -N2- ((S) -1-hydroxybut-3-en-2-yl) -4-oxo-1, 4-dihydropyridine-2, 5-dicarboxamide (300 mg,0.543 mmol) was dissolved in acetonitrile (1 mL). The resulting mixture was heated to 88 ℃. To the hot mixture was added paraformaldehyde (42.9 mg,1.36 mmol), followed by TFA (0.15 mL). Heating was continued for 15 hours. The reaction was then cooled to room temperature and concentrated. The resulting residue was then dissolved in DMF (2 mL) and benzyl bromide (111 mg,0.652 mmol) and potassium carbonate (600 mg,4.34 mmol) were added sequentially. The resulting mixture was heated to 70 ℃ for 2 hours. The resulting mixture was then cooled to room temperature, partitioned between ethyl acetate and water, the organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated, and purified by normal phase chromatography. LCMS-esi+ (m/z): h+ calculated theory for C30H30F2N4O 5: 564.22, found: 565.102.
Step 6: synthesis of 5- (benzyloxy) -1- (but-3-en-2-yl) -N- (2, 4-difluorobenzyl) -3- ((S) -1-fluorobutyryl- 3-en-2-yl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
5- (benzyloxy) -1- (but-3-en-2-yl) -N- (2, 4-difluorobenzyl) -3- ((S) -1-hydroxybut-3-en-2-yl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (180 mg,0.32 mmol) was dissolved in DCM (3 mL) and cooled to 0 ℃. To the cold mixture was added dropwise bis (2-methoxyethyl) aminothiotrifluoride (423 mg,1.91 mmol). After the addition, the reaction was taken out of the cooling bath and allowed to warm to room temperature and stirred at this temperature overnight. The reaction was then cooled back to 0 ℃, and quenched dropwise with saturated sodium bicarbonate with vigorous stirring. The mixture was extracted with DCM. The organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated and purified by normal phase chromatography. LCMS-esi+ (m/z): h+ calculated theory for C30H29F3N4O 4: 566.21, found: 567.120.
step 7: synthesizing (1R, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -5- (fluoromethyl) -2-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
5- (benzyloxy) -1- (but-3-en-2-yl) -N- (2, 4-difluorobenzyl) -3- ((S) -1-fluorobut-3-en-2-yl) -4, 6-dioxo-2, 3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (100 mg,0.177 mmol) was dissolved in DCE (3 mL). Hoveyda-Grubbs II catalyst (11.1 mg,0.0177 mmol) was added. The resulting mixture was purged with argon for 5 minutes, then sealed and heated at 80 ℃ overnight. The reaction was cooled to room temperature, concentrated and purified by normal phase chromatography. LCMS-esi+ (m/z): h+ calculated theory for C28H25F3N4O 4: 538.18, found: 539.105.
step 8: synthesis of (1R, 2S, 5S) -N- (2, 4-difluorobenzyl) -5- (fluoromethyl) -8-hydroxy-2-methyl-7, 9-di- Oxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide
(1R, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -5- (fluoromethyl) -2-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (20 mg,0.0371 mmol) was dissolved in EtOH (20 mL) at room temperature. To the stirred mixture was added 10% Pd/C (4 mg). The resulting mixture was degassed and flushed three times with nitrogen, degassed and flushed three times with hydrogen, and then hydrogenated under a hydrogen balloon for 1 hour. The reaction was degassed and flushed with nitrogen and filtered through celite. The filtrate was concentrated and purified by reverse phase prep HPLC. LCMS-esi+ (m/z): h+ calculated theory for C21H21F3N4O 4: 450.15, found: 451.224.1H NMR (400 MHz, DMSO-d 6) δ10.34 (t, J=5.9 Hz, 1H), 8.34 (s, 1H), 7.42 (td, J=8.6, 6.6Hz, 1H), 7.25 (ddd, J=10.5, 9.3,2.6Hz, 1H), 7.07 (td, J=8.6, 2.6Hz, 1H), 4.74 (s, 2H), 4.71-4.43 (m, 6H), 1.94-1.81 (m, 1H), 1.77-1.65 (m, 1H), 1.61-1.39 (m, 2H), 1.28 (d, J=7.1 Hz, 3H).
Step 97: preparation of (1R, 2S, 5S) -N- (2, 4-difluorobenzyl) -5- (fluoromethyl) -8-hydroxy-2-methyl-7, 9- Dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (C97)
(1R, 2S, 5S) -8- (benzyloxy) -N- (2, 4-difluorobenzyl) -5- (fluoromethyl) -2-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (20 mg,0.0371 mmol) prepared according to example 96 was treated with a mixture of DCM (1.5 mL) and TFA (1.5 mL) at room temperature for 2 hours. The reaction was concentrated and purified by reverse phase prep HPLC. LCMS-esi+ (m/z): h+ calculated theory for C21H19F3N4O 4: 448.14, found: 449.179.1H NMR (400 MHz, acetonitrile-d 3) delta 10.19-10.11 (m, 1H), 8.39 (s, 1H), 7.50-7.38 (m, 1H), 7.02-6.92 (m, 2H), 5.81 (dt, J=11.4, 2.3Hz, 1H), 5.66-5.59 (m, 1H), 5.54-5.39 (m, 1H), 5.13 (d, J=14.6 Hz, 1H), 4.80-4.55 (m, 5H), 3.92-3.82 (m, 1H), 1.37 (d, J=6.7 Hz, 3H).
Example 98: preparation of (1S, 2R, 5R) -N- (2, 4-difluorobenzyl) -5- (fluoromethyl) -8-hydroxy-2-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-A Amide (C98)
(1S, 2R, 5R) -N- (2, 4-difluorobenzyl) -5- (fluoromethyl) -8-hydroxy-2-methyl-7, 9-dioxo-2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide was prepared according to example 96, except that (2R) -2-aminobut-3-en-1-ol was used instead of (2S) -2-aminobut-3-en-1-ol in step 3. LCMS-esi+ (m/z): h+ calculated theory for C21H21F3N4O 4: 450.15, found: 451.12.1H NMR (400 MHz, acetonitrile-d 3) δ10.33 (s, 1H), 8.45 (s, 1H), 7.44 (td, J=9.2, 8.8,6.5Hz, 1H), 7.06-6.86 (m, 2H), 4.79-4.39 (m, 7H), 3.63-3.58 (m, 1H), 1.95-1.82 (m, 2H), 1.62 (dt, J=7.1, 3.5Hz, 2H), 1.32 (d, J=7.2 Hz, 3H).
Example 99: preparation of (1S, 2R,4S, 5S) -4-fluoro-5- (fluoromethyl) -8-hydroxy-2-methyl-7, 9-dioxo-) N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclic ring Nonane-10-carboxamide (C99)
Step 1: synthesis of (1S, 2R,4R, 5R) -8- (benzyloxy) -5- (fluoromethyl) -4-hydroxy-2-methyl-7, 9-dioxo substituted-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triaza-s Cyclononane-10-carboxamide
To (1S, 2R, 5R) -8- (benzyloxy) -5- (fluoromethyl) -2-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 50 in IPA (3 ml)][1,2,5]Triazacyclononane-10-carboxamide (116 mg,0.208 mmol) to which PhSiH was added 3 (45.1 mg,0.417 mmol) followed by the addition of the Shenvi catalyst (7.56 mg,0.0012 mmol) and then the mixture was taken up in O 2 Stirred at room temperature for 24 hours. The reaction was quenched by addition of 10% sodium thiosulfate, which was then extracted with EtOAc. The organic phase was washed with brine and with MgSO 4 And (5) drying. The solvent was removed under vacuum. The crude material was purified by column on silica gel to give the title compound. MS (M/z) [ M+H ]] + 574.98。
Step 2: synthesis of (1S, 2R,4S, 5S) -8- (benzyloxy) -4-fluoro-5- (fluoromethyl) 2-methyl-7, 9-dioxo-) N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclic ring Nonane-10-carboxamide
(1S, 2R,4R, 5R) -8- (benzyloxy) -5- (fluoromethyl) -4-hydroxy-2-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] in a plastic vial at 0 DEG C][1,2,5]To triazacyclononane-10-carboxamide (30 mg,0.0522 mmol) was added deoxofluor (2.7N in toluene) in DCM (1 mL) (0.193 mL,0.522 mmol). The reaction was then stirred at room temperature overnight. Saturated NaHCO for reaction 3 Quench at 0 ℃ and extract three times with DCM. The organic phase was washed with brine and with MgSO 4 And (5) drying. The crude material was purified by column on silica gel to give the title compound. MS (M/z) [ M+H ]] + 577.4。
Step 3: synthesis of (1S, 2R,4S, 5S) -4-fluoro-5- (fluoromethyl) -8-hydroxy-2-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclonones Alkane-10-carboxamides
To (1S, 2R,4S, 5S) -8- (benzyloxy) -4-fluoro-5- (fluoromethyl) -2-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ] at room temperature][1,2,5]Triazacyclononane-10-carboxamide (66.2 mg,0.115 mmol) TFA (0.5 mL) in toluene (0.5 mL) was added. The reaction mixture was stirred at room temperature overnight. The solvent was removed in vacuo and the crude material was purified by prep HPLC to give the title compound. MS (M/z) [ M+H ] ] + 487.05.1H NMR (400 MHz, chloroform-d) δ10.18 (d, J=5.1 Hz, 1H), 8.59 (s, 1H), 6.80-6.56 (m, 2H), 5.41-5.21 (m, 1H), 5.12-4.88 (m, 2H), 4.86-4.55 (m, 4H), 3.49 (s, 1H), 2.46-2.26 (m, 1H), 2.10-1.86 (m, 1H), 1.56 (dd, J=7.3, 2.3Hz, 3H), 1.54-1.14 (m, 1H).
Example 100: preparation of (1S, 2R, 5S) -8-hydroxy-2, 5, 13-trimethyl-7, 9-dioxo-N- (2, 4, 6-trio Fluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C100)
Step 1: synthesis of 1- ((R) -but-3-en-2-yl) -3- ((S) -but-3-en-2-yl) -5-hydroxy-2-methyl-4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxylic acid Amines
To a 20mL microwave vial was added sequentially in a manner similar to example 24 except that 3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -1- (((R) -but-3-en-2-yl) amino) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) -1, 4-dihydropyridine-2, 5-dicarboxamide (500 mg,0.90mmol,1.0 eq), acetaldehyde (0.5 mL,8.97mmol,10 eq), dichloroethane (3.2 mL) and acetonitrile (3.2 mL), glacial acetic acid (354 μL,6.14mmol,6.8 eq) and trifluoroacetic acid (354 μL,4.58mmol,5.1 eq) prepared using (S) -but-3-en-2-ol in place of but-3-en-2-ol in step 2, and the vial was sealed and transferred immediately to a hot plate preheated to 90 ℃. The solution was stirred at 90 ℃ for 16.5 hours, then the vial was removed from the heat source and allowed to cool to ambient temperature. The contents of the vials were combined in a 100mL Erlenmeyer flask (Erlenmeyer flask), saturated sodium bicarbonate (30 mL) and ethyl acetate (20 mL) were added and stirred for 30 min. The layers were then separated, the aqueous layer was extracted into ethyl acetate (2×20 mL), and the combined organic extracts were dried over magnesium sulfate, filtered and concentrated. The resulting residue was purified by flash column chromatography (0% -100% etoac/hexanes) to give 1- ((R) -but-3-en-2-yl) -3- ((S) -but-3-en-2-yl) -5-hydroxy-2-methyl-4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide. ES/MS 491.171[ M+H ] +.
Step 2: synthesis of 5- (benzyloxy) -1- ((R) -but-3-en-2-yl) -3- ((S) -but-3-en-2-yl) -2-methyl 1-hydroxy-4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7- Formamide
1- ((R) -but-3-en-2-yl) -3- ((S) -but-3-en-2-yl) -5-hydroxy-2-methyl-4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] in a 20mL vial][1,2,4]Triazine-7-carboxamide (123 mg,0.25mmol,1.0 eq.) potassium carbonate (178 mg,1.3mmol,5.1 eq.) and dimethylformamide (6 mL). Benzyl bromide (89 μl,0.75mmol,3.0 eq.) was added to the suspension, then the suspension was heated to 90 ℃ for 23 hours, then the reaction was removed from the heating and allowed to cool to ambient temperature. The reaction mixture was quenched with aqueous lithium chloride (10% w/w,10 mL) and the layers separated. Extracting the aqueous layer to CH 2 Cl 2 (2X 15 mL) and the combined organic extracts were washed with water (2X 15 mL), brine (15 mL), dried over magnesium sulfate, filtered and concentrated. The resulting residue was purified by flash column chromatography (0% -100% etoac/hexanes) to give 5- (benzyloxy) -1- ((R) -but-3-en-2-yl) -3- ((S) -but-3-en-2-yl) -2-methyl-4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ][1,2,4]Triazine-7-carboxamide. ES/MS 581.182[ M+H ]]+。
Step 3: synthesis of (1S, 2R, 5S) -8- (benzyloxy) -2,5, 13-trimethyl-7, 9-dioxo-N- (2, 4, 6-trio Fluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide
To a 20mL vial was added 5- (benzyloxy) -1- ((R) -but-3-en-2-yl) -3- ((S) -but-3-en-2-yl) -2-methyl-4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (76 mg,0.13mmol,1.0 eq.), a second generation Hoyveda-Grubbs catalyst (8.3 mg,0.013mmol,10 mol%) and dichloroethane (5 mL). The solution was degassed and placed under an argon atmosphere (this was done in triplicate). The flask was then equipped with an air condenser and heated at 80 ℃ for 21 hours, after which the flask was removed from the heating and allowed to cool to ambient temperature. The solids were removed by filtration, the volatiles were removed in vacuo and the resulting residue was purified by flash column chromatography (0% -100% etoac/hexanes) to give (1 s,2r,5 s) -8- (benzyloxy) -2,5, 13-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide. ES/MS 553.183[ M+H ] +.
Step 4: synthesis of (1S, 2R, 5S) -8-hydroxy-2, 5, 13-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide
To a 1-dram vial was added (1S, 2R, 5S) -8- (benzyloxy) -2,5, 13-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (43 mg,0.077mmol,1.0 eq.), toluene (0.7 mL) and trifluoroacetic acid (0.7 mL). The reaction was stirred at ambient temperature for 4.5 hours, then the volatiles were removed in vacuo and the resulting residue was purified by preparative HPLC (0% -100% ch with 0.1% tfa modifier 3 CN/H 2 O) to give (1S, 2R, 5S) -8-hydroxy-2, 5, 13-trimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide. ES/MS 463.215[ M+H ]]+。 1 H NMR (400 MHz, chloroform-d) δ10.25 (s, 1H), 8.50 (d, j=1.1 hz, 1H), 6.67 (t, j=8.1 hz, 2H), 5.59 (d, j=11.5 hz, 1H), 5.50-5.39 (m, 1H), 5.35 (dt, j=11.6, 3.0hz, 1H), 5.18 (q, j=6.7 hz, 1H), 4.75-4.59 (m, 2H), 3.66 (dt, j=6.8, 3.3hz, 1H), 1.38 (dd, j=7.1, 5.9hz, 6H), 1.28 (d, j=6.6 hz, 3H).
Example 101: preparation of (1S, 2R, 5S) -13-ethyl-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxylic acid Amine (C101)
(1S, 2R, 5S) -13-ethyl-8-hydroxy-2, 5-dimethyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide was prepared in a similar manner to example 100 except that propionaldehyde was used instead of acetaldehyde in step 1And outside. ES/MS 477.186[ M+H ]]+。 1 H NMR (400 MHz, chloroform-d) delta 10.24 (s, 1H), 8.49 (s, 1H), 6.67 (t, j=8.1 hz, 2H), 5.66-5.50 (m, 1H), 5.50-5.42 (m, 1H), 5.34 (dt, j=11.6, 3.0hz, 1H), 4.83 (t, j=7.3 hz, 1H), 4.74-4.58 (m, 2H), 3.68 (dd, j=6.6, 3.3hz, 1H), 1.52 (ddp, j=21.9, 14.5,7.5hz, 2H), 1.38 (dd, j=10.5, 7.1hz, 6H), 0.96 (t, j=7.4 hz, 3H).
Example 102: preparation of (1S, 2R, 5S) -N- ((4-chloro-3, 5-difluoropyridin-2-yl) methyl) -8-hydroxy-2, 5- Dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide (C102)
Step 1: synthesis of (4-chloro-3, 5-difluoropyridin-2-yl) methylamine
Will beMolecular sieves (1 g) added to 4mL 7N NH 3 In MeOH. Stirring is carried out for 2 hours. The molecular sieve was removed by filtration. The filtrate was treated with 4-chloro-3, 5-difluoropyridine-carbaldehyde (202 mg,1.14 mmol) at room temperature. The reaction mixture was stirred at room temperature overnight. The reaction mixture was cooled to 0 ℃. NaBH addition at 0deg.C 4 (65 mg,1.71 mmol). The reaction mixture was then warmed to room temperature. The reaction was saturated with NaHCO over 2 hours 3 (10 mL) quenching. EtOAc (2×10 mL) was added to extract the crude product. The organic layer was then treated with 1N HCl (10 mL). The aqueous layer was collected and treated with NaHCO 3 The (saturation) treatment caused ph=8. The product was extracted with Me-THF (1X 10 mL). The organic phase was separated and concentrated to dryness to give the product which was used directly without further purification. MS (m/z): 179.0[ M+H ]]+。
Step 2: synthesis of (1S, 2R, 5S) -8- (benzyloxy) -N- ((4-chloro-3, 5-difluoropyridin-2-yl) methyl) -2,5- Dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2 ]-b][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 63 was reacted at room temperature][1,2,5]Triazacyclononane-10-carboxylic acid (150 mg,0.379 mmol) was dissolved in DMF (1 mL). DIEA (0.264 mL,1.52 mmol) was added under an argon atmosphere. The resulting reaction mixture was cooled to 0 ℃. HATU (216 mg,0.569 mmol) was then added. The resulting reaction mixture was then warmed to room temperature and stirred at room temperature for 1 hour. To the reaction mixture was added a solution of (3-chloro-2, 4-difluorophenyl) methylamine (102 mg,0.569 mmol) in DMF (0.5 mL). The reaction mixture was then stirred at room temperature for 17 hours. The reaction mixture was diluted with EtOAc (10 mL) and saturated NH 4 A mixture of aqueous Cl (10 mL) and water (10 mL) was treated. The organic phase was then washed with water (10 mL) followed by saturated brine (10 mL). The organic phase was then separated and concentrated. The residue was purified on a silica gel column with 0% -100% etoac/Hex to give the product. MS (m/z): 556.1[ M+H ]]+。
Step 3: synthesis of (1S, 2R, 5S) -N- ((4-chloro-3, 5-difluoropyridin-2-yl) methyl) -8-hydroxy-2, 5-dimethyl 1, 2-b-methyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido][1,2,5]Triazacyclononane-10-A Amides and their use
(1S, 2R, 5S) -8- (benzyloxy) -N- ((4-chloro-3, 5-difluoropyridin-2-yl) methyl) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] at room temperature][1,2,5]Triazacyclononane-10-carboxamide (100 mg,0.180 mmol) was dissolved in toluene (2 mL). TFA (2 mL) was carefully added with stirring. The resulting reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was then concentrated to dryness. The residue was dissolved in MeOH and taken up in 0.1% TFA in 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilizing to obtain the final product. MS (m/z): 466.2[ M+H ]]+; 1 H NMR (400 MHz, acetonitrile-d 3) delta 10.43 (s, 1H), 8.44 (s, 1H), 8.39 (s, 1H), 5.67 (dt, j=11.4, 2.5hz, 1H), 5.40 (ddt, j=16.9, 10.3,5.3hz, 2H), 5.02 (d, j=14.3 hz, 1H), 4.78 (dd, j=5.7, 1.8hz, 2H),4.58(d,J=14.3Hz,1H),3.84(tt,J=6.7,3.4Hz,1H),1.35(dd,J=7.1,2.9Hz,6H)。
Example 103: preparation of (1S, 2R, 5S) -N- (3-chloro-2, 4, 6-trifluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide (C103)
Step 1: synthesis of (3-chloro-2, 4, 6-trifluorophenyl) methylamine
Will beMolecular sieves (1 g) added to 4mL 7N NH 3 In MeOH. Stirring is carried out for 2 hours. The molecular sieve was removed by filtration. The filtrate was treated with 3-chloro-2, 4, 6-trifluorobenzaldehyde (1000 mg,5.14 mmol) at room temperature. The reaction mixture was stirred at room temperature overnight. The reaction mixture was cooled to 0 ℃. NaBH addition at 0deg.C 4 (292 mg,7.71 mmol). The reaction mixture was then warmed to room temperature. The reaction was saturated with NaHCO over 2 hours 3 (10 mL) quenching. EtOAc (2×10 mL) was added to extract the crude product. The organic layer was then treated with 1N HCl (10 mL). The aqueous layer was collected and treated with NaHCO 3 The (saturation) treatment caused ph=8. The product was extracted with Me-THF (1X 10 mL). The organic phase was separated and concentrated to dryness to give the product which was used directly without further purification. MS (m/z): 196.0[ M+H ]]+。
Step 2: synthesizing (1S, 2R, 5S) -8- (benzyloxy) -N- (3-chloro-2, 4, 6-trifluoro-benzyl) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] ][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] prepared according to example 63 was reacted at room temperature][1,2,5]Triazacyclononane-10-carboxylic acid (200 mg,0.506 mmol) was dissolved in DMF (1 mL). DIEA (0.35 mL,2.02 mmol) was added under an argon atmosphere. The resulting reaction mixture was cooled to 0 ℃. HATU (250 mg, 0.618 mmol) was then added. The resulting reaction mixture was then warmed to room temperature and stirred at room temperature for 1 hour. To the reaction mixture was added a solution of (3-chloro-2, 4, 6-trifluorophenyl) methylamine (148 mg,0.759 mmol) in DMF (0.5 mL). The reaction mixture was then stirred at room temperature for 17 hours. The reaction mixture was diluted with EtOAc (10 mL) and saturated NH 4 A mixture of aqueous Cl (10 mL) and water (10 mL) was treated. The organic phase was then washed with water (10 mL) followed by saturated brine (10 mL). The organic phase was then separated and concentrated. The residue was purified on a silica gel column with 0% -100% etoac/Hex to give the product. MS (m/z): 572.9[ M+H ]]+。
Step 3: synthesis of (1S, 2R, 5S) -N- (3-chloro-2, 4, 6-trifluorobenzyl) -8-hydroxy-2, 5-dimethyl-7, 9-dio Oxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] ][1,2,5]Triazacyclononane-10-carboxamide
(1S, 2R, 5S) -8- (benzyloxy) -N- (3-chloro-2, 4, 6-trifluorobenzyl) -2, 5-dimethyl-7, 9-dioxo-2, 5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] at room temperature][1,2,5]Triazacyclononane-10-carboxamide (130 mg,0.227 mmol) was dissolved in DMF (2 mL). LiCl (58 mg,1.36 mmol) was added. The resulting reaction mixture was heated at 100℃with stirring for 6 hours. The reaction mixture was then diluted with EtOAc (10 mL) and combined with NH 4 Cl (saturated aqueous) (10 mL) and 1N HCl (10 mL). The organic phase was separated and washed with water (10 mL) and brine (10 mL). The organic extract was concentrated to give the crude product. Further purification on a silica gel column (0% -100% EtOAc/Hex) afforded the product. MS (m/z): 483.2[ M+H ]]++, of the material; 1H NMR (400 MHz, acetonitrile-d 3) δ10.26 (s, 1H), 8.37 (s, 1H), 7.05 (td, J=9.5, 2.2Hz, 1H), 5.66 (dt, J=11.4, 2.4Hz, 1H), 5.46-5.32 (m, 2H), 5.01 (d, J=14.4 Hz, 1H), 4.65 (d, J=6.0 Hz, 2H), 4.56 (d, J=14.4 Hz, 1H), 3.83 (tt, J=7.0, 3.6Hz, 1H), 1.37-1.28 (m, 6H).
Example 104: preparation of (1 'S,5' S) -8 '-hydroxy-5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) Radical) -7',9' -dihydro-5 'H-spiro [ cyclobutane-1, 2' - [1,6 ]]Methanopyrido [1,2-b ] bridged][1,2,5]Triazacyclonones Alkyl (C)]-10' -carboxamide (C104)
Step 1: synthesis of di-tert-butyl 1- (1-formylcyclobutyl) hydrazine-1, 2-dicarboxylate
Ring Ding Jiaquan (1.64 g,19.5 mmol) and di-tert-butyl-diazene-1, 2-dicarboxylic acid ester (3 g,13 mmol) were dissolved in DCM. L-proline (0.15 g,1.3 mmol) was then added. The slurry was stirred at room temperature for 2 hours and then heated at 40 ℃ for 5 hours. The solvent was removed by rotary evaporation. The residue was partitioned between EtOAc (50 mL) and water (50 mL). Brine was added to facilitate separation. The organic phase was separated and washed with water and brine. The organic phase was separated and concentrated to give a pale yellow solid. Purification on a silica gel column with 0% -100% etoac in hexanes afforded the product. 1H NMR (400 MHz, CD3 CN) δ9.99-9.39 (br, 1H), 7.69-6.55 (br, 1H), 2.48-2.20 (m, 4H), 1.82 (d, J=15.2 Hz, 2H), 1.61-1.34 (m, 18H).
Step 2: synthesis of di-tert-butyl 1- (1-vinylcyclobutyl) hydrazine-1, 2-dicarboxylate
KOTBu (1.51 g,13.5 mmol) was added to a suspension of methyltriphenyl phosphonium iodide (5.56 g,13.5 mmol) in THF (25 mL) at 0deg.C. The mixture was stirred for 30 minutes. A solution of di-tert-butyl 1- (1-formylcyclobutyl) hydrazine-1, 2-dicarboxylate (2.12 g,6.74 mmol) in THF (10 mL) was added dropwise over 10 min. The reaction mixture was warmed to room temperature. After stirring for 17 hours, the reaction mixture was cooled to 0 ℃. EtOAc (20 mL) was added. The resulting reaction mixture was treated with saturated aqueous NH4Cl (10 mL) and water (10 mL). The organic phase was separated and the aqueous phase was extracted with EtOAc (2X 10 mL). The combined organic phases were concentrated. Purification by column chromatography on silica gel (0% -50% etoac/hexanes) afforded the product. 1H NMR (400 MHz, CD3 CN) delta 7.01 (br, 1H), 6.14 (dd, J=17.4, 10.6Hz, 1H), 5.31-4.97 (m, 2H), 2.53-2.22 (m, 2H), 2.17-1.99 (m, 2H), 1.83-1.64 (m, 2H), 1.44 (d, J=16.0 Hz, 18H).
Step 3: synthesis of 2- (1-vinylcyclobutyl) hydrazine-1-onium 2, 2-trifluoroacetate
Di-tert-butyl 1- (1-vinylcyclobutyl) hydrazine-1, 2-dicarboxylate (1.25 g,4 mmol) was dissolved in DCM (15 mL) at room temperature and cooled with an ice-water bath under argon. TFA (15 mL) was added. The resulting reaction mixture was allowed to warm to room temperature and stirred for 17 hours. The reaction mixture was then concentrated to dryness. The residue was placed under high vacuum for 5 hours to give the product. MS (m/z): 113.0[ M+H ] +.
Step 4: synthesis of 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1- ((1-ethylene Methyl (phenylcyclobutyl) amino) -1, 4-dihydropyridine-2-carboxylate
2- (1-vinylcyclobutyl) hydrazine-1-onium 2, 2-trifluoroacetate (900 mg,4 mmol) was dissolved in MeOH (20 mL). NaHCO3 (1.4 g,16.8 mmol) was then added as a solid. 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -4H-pyran-2-carboxylic acid methyl ester (1.5 g,3.35 mmol) was then added in one portion as a solid. Water (10 mL) was added. The resulting reaction mixture was heated at 55 ℃ for 17 hours. The reaction mixture was then diluted with EtOAc (20 mL) and treated with brine (20 mL) and water (20 mL). The organic phase was separated and concentrated to give the product. MS (m/z): 542.0[ M+H ] +.
Step 5: synthesis of 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1- ((1-ethylene Phenylcyclobutyl) amino) -1, 4-dihydropyridine-2-carboxylic acid
Methyl 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1- ((1-vinylcyclobutyl) amino) -1, 4-dihydropyridine-2-carboxylate (1.82 g,3.36 mmol) was dissolved in MeOH (30 mL), water (15 mL) and THF (10 mL) at room temperature. LiOH (5M aqueous solution, 5.38 mL) was added. The reaction mixture was stirred at room temperature for 20 hours. The reaction mixture was carefully concentrated to remove MeOH. The residue was diluted and rinsed with some water (30 mL) and acidified with 1N HCl to ph=3. EtOAc (50 mL) was added for extraction. The organic phase was separated. The aqueous layer was extracted with more EtOAc (30 mL). The combined organic phases were washed with water and brine. The separated organic phase is passed throughNa 2 SO 4 Drying, filtering and concentrating to obtain an acidic product. MS (m/z): 527.9[ M+H ]]+。
Step 6: synthesis of (S) -3- (benzyloxy) -N2- (but-3-en-2-yl) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) Phenyl) -1- ((1-vinylcyclobutyl) amino) -1, 4-dihydropyridine-2, 5-dicarboxamide
3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1- ((1-vinylcyclobutyl) amino) -1, 4-dihydropyridine-2-carboxylic acid (1.71 g,3.24 mmol) was dissolved in DMF (20 mL) at room temperature under argon. DIEA (2.82 mL,16.2 mmol) was added. The reaction mixture was cooled with an ice-water bath. HATU (1.946 g,4.86 mmol) was then added. The reaction mixture was stirred at low temperature for 5 minutes, then the cold bath was removed. The reaction mixture was stirred at room temperature for 1 hour. (S) -but-3-en-2-amine HCl salt (0.816 g,7.59 mmol) was then added in one portion at room temperature. The reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was then diluted with EtOAc (50 mL) and sequentially with NaHCO 3 (saturated) (20 mL)/water (20 mL), NH4Cl (20 mL), water (20 mL), and brine (20 mL). The organic phase was taken up in Na 2 SO 4 Drying and filtering. Concentrating the filtrate to obtain the product. MS (m/z): 581.0[ M+H ]]+。
Step 7: synthesis of (S) -3- (but-3-en-2-yl) -5-hydroxy-4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) scheme 1- (1-vinylcyclobutyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
(S) -3- (benzyloxy) -N2- (but-3-en-2-yl) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) -1- ((1-vinylcyclobutyl) amino) -1, 4-dihydropyridine-2, 5-dicarboxamide (1.8 g,3.1 mmol) was dissolved in ACN (6 mL) and DCE (6 mL). Paraformaldehyde (248 mg,7.75 mmol) was added. AcOH (0.86 mL) and TFA (0.86 mL) were added dropwise in this order without stirring. The reaction was heated to 85 ℃ under argon with stirring and maintained under this condition for 24 hours. The reaction mixture was then concentrated to dryness. The residue was purified on a silica gel column with 0% -100% etoac/Hex to give the product. MS (m/z): 503.2[ M+H ] +.
Step 8: synthesis of (S) -5- (benzyloxy)) -3- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) 1- (1-vinylcyclobutyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxamide
(S) -3- (but-3-en-2-yl) -5-hydroxy-4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -1- (1-vinylcyclobutyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] at room temperature ][1,2,4]Triazine-7-carboxamide (1.16 g,2.31 mmol) was dissolved in DMF (5 mL). Adding K 2 CO 3 (2.44 g,17.7 mmol). BnBr (1.38 g,8.08 mmol) was then added. The reaction mixture was heated to 73 ℃ (bath temperature) for 5 hours. The reaction mixture was diluted with EtOAc (50 mL) and treated with water (20 mL). The organic phase was separated. The aqueous layer was extracted with EtOAc (20 mL). The combined organic phases were washed with water and brine. The organic phase was concentrated to give the crude product. Further purification on a silica gel column with 0% -100% etoac/Hex afforded the product. MS (m/z): 593.1[ M+H ]]+。
Step 9: synthesis of (1 'S,5' S) -8'- (benzyloxy) -5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) Radical) -7',9' -dihydro-5 'H-spiro [ cyclobutane-1, 2' - [1,6 ]]Methanopyrido [1,2-b ] bridged][1,2,5]Triazacyclonones Alkyl (C)]-10' -carboxamide
(S) -5- (benzyloxy) -3- (but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -1- (1-vinylcyclobutyl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (0.5 g,0.85 mmol) was dissolved in dichloroethane (15 mL). The solution was bubbled with argon for 10 minutes. Grubbs catalyst M202 (211 mg,0.22 mmol) was added. Argon purge was then applied again for 10 minutes. The reaction mixture was heated at 80 ℃ under argon for 24 hours. Another round of Grubbs M202 (211 mg, 0.22) addition was applied followed by a 10 minute purge with argon. The reaction mixture was again heated at 80 ℃ for 24 hours. The reaction mixture was then concentrated to dryness. The residue was purified on a silica gel column with 0% -100% etoac/Hex to give the product. MS (m/z): 565.1[ M+H ] +.
Step 10: synthesis of (1 'S,5' S) -8 '-hydroxy-5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) scheme 7',9' -dihydro-5 'H-spiro [ cyclobutane-1, 2' - [1,6 ]]Bridge sub-systemMethylpyrido [1,2-b ]][1,2,5]Triazacyclononane]- 10' -carboxamide
(1 'S,5' S) -8'- (benzyloxy) -5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) -7',9' -dihydro-5 'H-spiro [ cyclobutane-1, 2' - [1,6 ]]Methanopyrido [1,2-b ] bridged][1,2,5]Triazacyclononane]10' -carboxamide (12 mg,0.0213 mmol) was dissolved in toluene (8 mL). TFA (9 mL) was added at room temperature. The resulting reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was then concentrated to dryness. The residue was treated with 0.1% TFA in the range of 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilizing to obtain the final product. MS (m/z): 475.2[ M+H ]]+。 1 H NMR(400MHz,CD3CN)δ10.23(s,1H),8.48(s,1H),7.05–6.82(m,2H),6.05(dd,J=11.8,2.6Hz,1H),5.64(dd,J=11.8,2.3Hz,1H),5.43–5.23(m,1H),5.00(d,J=14.4Hz,1H),4.73–4.59(m,2H),4.55(d,J=14.4Hz,1H),2.45–2.32(m,1H),2.32–2.22(m,1H),1.86–1.57(m,4H),1.32(d,J=7.3Hz,3H)。
Example 105: preparation of (1 'S,5' S) -8 '-hydroxy-5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) Phenyl) -4',5',7',9' -tetrahydro-3 'h-spiro [ cyclobutane-1, 2' - [1,6 ]]Methanopyrido [1,2-b ] bridged][1,2,5]Trinitrogen Heterocyclononane]-10' -carboxamide (C105)
(1 'S,5' S) -8'- (benzyloxy) -5' -methyl-7 ',9' -dioxo-N- (2, 4, 6-trifluorobenzyl) -7',9' -dihydro-5 'H-spiro [ cyclobutane-1, 2' - [1,6 ] prepared according to example 104 ]Methanopyrido [1,2-b ] bridged][1,2,5]Triazacyclononane]-10' -carboxamide (10 mg,0.0177 mmol) was dissolved in MeOH (10 mL). Pd/C (10%) (12 mg) was added. The reaction mixture was taken up in H 2 Stirring is carried out for 6 hours under a balloon atmosphere. The reaction mixture was then filtered to remove the catalyst. The filtrate was concentrated to dryness. The residue was treated with 0.1% TFA in the range of 0-100% CH 3 The aqueous CN solution was purified by reverse phase prep HPLC to give the desired product. Lyophilizing to obtain the final product. MS (m/z): 477.1[ M+H ]]+。1H NMR(400MHz,CD3CN)δ10.35(s,1H),8.50(s,1H),6.87(t,J=8.5Hz,2H),4.64–4.57(m,4H),4.45(d,J=14.6Hz,1H),2.21–2.00(m,4H),1.85(dt,J=15.4,5.0Hz,2H),1.74-1.39(m,4H),1.23(d,J=6.7Hz,3H)。
Example 106: preparation of (1S, 2S, 5S) -2-ethyl-8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trio Fluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane-10-carboxamide (C106)
(1S, 2S, 5S) -8- (benzyloxy) -2-ethyl-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-B ] prepared according to example 72 step 2 as secondary product (B)][1,2,5]A reaction mixture of triazacyclononane-10-carboxamide (8 mg,0.0145 mmol) and lithium chloride (6.1 mg,0.145 mmol) in DMF (1 mL) was stirred overnight at 100deg.C. The reaction mixture was filtered, loaded onto reverse phase HPLC and eluted with 5% -100% aqueous acetonitrile to give the title product. MS (m/z) 463.20[ M+H ] ] + .1H NMR (400 MHz, methanol-d 4) delta 8.51 (s, 1H), 6.98-6.86 (m, 2H), 5.74 (ddd, J=12.3, 3.1,1.8Hz, 1H), 5.57 (ddd, J=12.4, 4.7,2.1Hz, 1H), 5.45-5.38 (m, 1H), 5.11 (d, J=14.4 Hz, 1H), 4.78 (d, J=14.4 Hz, 1H), 4.69 (d, J=2.3 Hz, 2H), 4.47 (dd, J=6.6, 3.1Hz, 1H), 1.61 (tt, J=12.3, 7.3Hz, 1H), 1.41 (d, J=7.3 Hz, 3H), 1.24 (ddd, J=13.3, 9.4, 7.82, 0.69 (j=3.3 Hz, 3H), 0.7.7 Hz, 3H).
Example 107: preparation of (1S, 2R, 5S) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) scheme 2- (2, 2-trifluoroethyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide (C107)
Step 1: synthesis of di-tert-butyl 1- (4, 4-trifluoro-1-oxobutan-2-yl) hydrazine-1, 2-dicarboxylate
4, 4-trifluorobutyraldehyde (2.46 g,19.5 mmol) and di-tert-butyl-diazene-1, 2-dicarboxylic acid ester (3 g,13 mmol) were dissolved in DCM (100 mL). L-proline (0.15 g,1.3 mmol) was then added. The slurry was stirred at room temperature for 2 hours and then heated at 40 ℃ for 17 hours. The solvent was removed. The residue was purified on a silica gel column with 0% -100% etoac in hexanes to give the product. 1H NMR (400 MHz, CD3 CN) δ9.66 (d, J=12.9 Hz, 1H), 7.70-7.37 (m, 1H), 4.67-4.62 (m, 1H), 2.87 (dtt, J=15.5, 12.0,5.8Hz, 1H), 2.63-2.44 (m, 1H), 1.51-1.40 (m, 18H).
Step 2: synthesis of di-tert-butyl 1- (5, 5-trifluoropent-1-en-3-yl) hydrazine-1, 2-dicarboxylate
To a suspension of methyltriphenyl phosphonium iodide (7.03 g,17 mmol) in THF (100 mL) at 0deg.C was added KOtBu (2.0 g,17.8 mmol). The mixture turned into a yellow suspension and was stirred for 30 minutes. A solution of di-tert-butyl 1- (4, 4-trifluoro-1-oxobutan-2-yl) hydrazine-1, 2-dicarboxylate (2.5 g,7.02 mmol) in THF (27 mL) was added dropwise over 10 min. The reaction mixture was warmed to room temperature. After stirring for 17 hours, the reaction mixture was cooled to 0deg.C and EtOAc (20 mL) was added. The resulting reaction mixture was treated with saturated aqueous NH4Cl (10 mL) and water (10 mL). The organic phase was separated and the aqueous phase was extracted with EtOAc (2X 10 mL). The combined organic phases were concentrated to give an orange foamy oil. Purification by column chromatography on silica gel (0% -50% etoac/hexanes) afforded the product. 1H NMR (400 MHz, CD3 CN) delta 7.31-6.47 (m, 1H), 6.05-5.78 (m, 1H), 5.38-5.12 (m, 2H), 4.94 (s, 1H), 2.83-2.36 (m, 2H), 1.45 (d, J=3.4 Hz, 18H).
Step 3: synthesis of (5, 5-trifluoropent-1-en-3-yl) hydrazinium trifluoroacetate salt
Di-tert-butyl 1- (5, 5-trifluoropent-1-en-3-yl) hydrazine-1, 2-dicarboxylate (1.76 g,4.97 mmol) was dissolved in DCM (40 mL) at room temperature and cooled with an ice-water bath under argon. TFA (20 mL) was added. The resulting reaction mixture was allowed to warm to room temperature and stirred for 17 hours. The reaction mixture was then concentrated to dryness and the residue was placed under high vacuum for 5 hours to give the product. MS (m/z) 155.1[ M+H ] +.
Step 4: synthesis of 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1- ((5, 5- Trifluoro-pent-1-en-3-yl) amino) -1, 4-dihydropyridine-2-carboxylic acid methyl ester
(5, 5-Trifluoropent-1-en-3-yl) hydrazine trifluoroacetate salt (1.32 g,4.96 mmol) was dissolved in MeOH (30 mL). NaHCO3 (3.474 g,41.4 mmol) was then added as a solid, some bubbles appeared. 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -4H-pyran-2-carboxylic acid methyl ester (1.85 g,4.14 mmol) was added in one portion as a solid. Water (15 mL) was added. The resulting reaction mixture was heated at 55 ℃ for 17 hours. The reaction mixture was then diluted with EtOAc (20 mL) and treated with brine (20 mL) and water (20 mL). The organic phase was separated and concentrated to give the product. MS (m/z) 584.3[ M+H ] +.
Step 5: synthesis of 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1- ((5, 5- Trifluoro-pent-1-en-3-yl) amino) -1, 4-dihydropyridine-2-carboxylic acid
Methyl 3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1- ((5, 5-trifluoropent-1-en-3-yl) amino) -1, 4-dihydropyridine-2-carboxylate (2.41 g,4.13 mmol) was dissolved in MeOH (30 mL), water (10 mL) and THF (15 mL) at room temperature. LiOH (5M in water) (6.61 mL,33.05 mmol) was added. The reaction mixture was stirred at room temperature for 20 hours. The reaction mixture was carefully concentrated to remove MeOH. The residue was diluted and rinsed with some water (30 mL) and then acidified with 1N HCl to ph=3. EtOAc (50 mL) was added and the organic phase was extracted with EtOAc (2×30 mL). The combined organic phases were washed with water and brine. The separated organic phase was dried over Na2SO4, filtered and concentrated to give the product. MS (m/z) 570.2[ M+H ] +.
Step 6: synthesis of 3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) 1- ((5, 5-trifluoropent-1-en-3-yl) amino) -1, 4-dihydropyridine-2, 5-dicarboxamide
3- (benzyloxy) -4-oxo-5- ((2, 4, 6-trifluorobenzyl) carbamoyl) -1- ((5, 5-trifluoropent-1-en-3-yl) amino) -1, 4-dihydropyridine-2-carboxylic acid (2.35 g,4.13 mmol) was dissolved in DMF (20 mL) at room temperature under argon. DIEA (3.59 ml,20.6 mmol) was added and the reaction mixture was cooled to 0 ℃. HATU (2.178 g,6.19 mmol) was then added and the reaction mixture was stirred at this temperature for 5 minutes before the cold bath was removed. The reaction mixture was stirred at room temperature for 1 hour. (S) -methylallylamine HCl salt (1.05 g,9.78 mmol) was then added in one portion at room temperature. The reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was then diluted with EtOAc (50 mL) and washed sequentially with NaHCO3 (saturated) (20 mL), water (20 mL), NH4Cl (20 mL), water (20 mL) and brine (20 mL). The organic phase was dried over Na2SO4 and filtered. Concentrating the filtrate to obtain the product. MS (m/z) 623.3[ M+H ] +.
Step 7: synthesis of 3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) 1- (((R) -5, 5-trifluoro-1-en-3-yl) amino) -1, 4-dihydropyridine-2, 5-dicarboxamide
A mixture of two diastereomers of 3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) -1- ((5, 5-trifluoropent-1-en-3-yl) amino) -1, 4-dihydropyridine-2, 5-dicarboxamide was chiral SFC with ADH-25 and eluted with IPA-NH3 to give 3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) -1- (((R) -5, 5-trifluoropent-1-en-3-yl) amino) -1, 4-dihydropyridine-2, 5-dicarboxamide as the first peak. MS (m/z) 623.3[ M+H ] +.
Step 8: synthesis of 3- ((S) -but-3-en-2-yl) -5-hydroxy-4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) scheme 1- ((R) -5, 5-Trifluoropent-1-en-3-yl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazine-7-carboxylic acid methyl ester Amides and their use
3- (benzyloxy) -N2- ((S) -but-3-en-2-yl) -4-oxo-N5- (2, 4, 6-trifluorobenzyl) -1- (((R) -5, 5-trifluoropent-1-en-3-yl) amino) -1, 4-dihydropyridine-2, 5-dicarboxamide (0.5 g,0.803 mmol) was dissolved in ACN (3 mL) and DCE (3 mL). Paraformaldehyde (63.4 mg,2.01 mmol) was added. AcOH (0.223 mL) and TFA (0.223 mL) were added dropwise without stirring. The reaction was heated to 85 ℃ under argon with stirring and maintained under this condition for 24 hours. The reaction mixture was then concentrated to dryness. The residue was purified on a silica gel column with 0% -100% etoac/Hex to give the product. MS (m/z) 545.1[ M+H ] +.
Step 9: synthesis of 5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) 1- ((R) -5, 5-Trifluoropent-1-en-3-yl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f][1,2,4]Triazines- 7-carboxamide
3- ((S) -but-3-en-2-yl) -5-hydroxy-4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -1- ((R) -5, 5-trifluoropent-1-en-3-yl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (70 mg,0.129 mmol) was dissolved in DMF (5 mL) at room temperature. K2CO3 (136 mg,0.98 mmol) was added. Benzyl bromide (77 mg,0.45 mmol) was then added. The reaction mixture was heated to 73 ℃ (bath temperature) for 5 hours. The reaction mixture was diluted with EtOAc (50 mL) and treated with water (20 mL). The organic phase was separated and the aqueous layer was extracted with EtOAc (20 mL). The combined organic phases were washed with water and brine. The organic phase was concentrated to give the crude product. Further purification on a silica gel column with 0% -100% etoac/Hex afforded the product. MS (m/z) 635.0[ M+H ] +.
Step 10: synthesis of (1S, 2R, 5S) -8- (benzyloxy) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl-) 2- (2, 2-trifluoroethyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide
5- (benzyloxy) -3- ((S) -but-3-en-2-yl) -4, 6-dioxo-N- (2, 4, 6-trifluorobenzyl) -1- ((R) -5, 5-trifluoropent-1-en-3-yl) -2,3,4, 6-tetrahydro-1H-pyrido [2,1-f ] [1,2,4] triazine-7-carboxamide (40 mg,0.063 mmol) was dissolved in dichloroethane (5 mL). Grubbs catalyst M720 (8.14 mg,0.0124 mmol) was added. Argon purge was then applied for 10 minutes. The reaction mixture was heated at 80 ℃ under argon for 24 hours. The reaction mixture was then concentrated to dryness. The residue was purified on a silica gel column with 0% -100% etoac/Hex to give the product. MS (m/z) 607.0[ M+H ] +.
Step 11: synthesis of (1S, 2R, 5S) -8-hydroxy-5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl-2-) (2, 2-trifluoroethyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b][1,2,5]Triazacyclononane (T-N-phenylmethane) 10-carboxamide
(1R, 2R, 5S) -8- (benzyloxy) -5-methyl-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2- (2, 2-trifluoroethyl) -2,5,7, 9-tetrahydro-1, 6-methanopyrido [1,2-b ] [1,2,5] triazacyclononane-10-carboxamide (38 mg,0.0627 mmol) was dissolved in DMF (3 mL). LiCl (34.2 mg, 0.803 mmol) was added at room temperature. The resulting reaction mixture was heated at 100℃with stirring for 7 hours. The reaction mixture was then diluted with EtOAc (10 mL) and then treated with 1N HCl (10 mL). The organic phase was separated and washed with water (10 mL) and brine (10 mL). The organic phase was concentrated to give a residue which was purified by reverse phase prep HPLC with 0% to 100% ch3cn in water with 0.1% tfa to give the desired product. Lyophilization gives the desired product. MS (m/z) 517.2[ M+H ] +.1H NMR (400 MHz, CD3 CN) δ10.18 (s, 1H), 8.43 (s, 1H), 6.88 (t, J=8.5 Hz, 2H), 5.79 (dt, J=11.8, 2.6Hz, 1H), 5.50 (dt, J=11.6, 3.1Hz, 1H), 5.38 (H, J=6.9 Hz, 1H), 4.98 (d, J=14.6 Hz, 1H), 4.71-4.56 (m, 3H), 4.17 (tq, J=6.1, 3.0Hz, 1H), 2.68 (ddd, J=15.8, 13.2,10.3,5.4Hz, 2H), 1.36 (d, J=7.4 Hz, 3H).
Examples 108 and 109: preparation of (1R) -8-hydroxy-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2, 3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2-b ]][1,2,5]Triazacyclononane-10-carboxamide and (1S) 8-hydroxy-7, 9-dioxo-N- (2, 4, 6-trifluorobenzyl) -2,3,4,5,7, 9-hexahydro-1, 6-methanopyrido [1,2- ] b][1,2,5]Triazacyclononane-10-carboxamide (C108 and C109)
Prepared according to example 2 of WO2019160783 using (2, 4, 6-trifluorophenyl) methylamine instead of (2, 4-difluorophenyl) methylamine in step 3. Separation by chiral SFC using 40% methanol gives peak 1 and peak 2.
Peak 1: MS-ESI (m/z) 423.1[ M+H ] +.1H NMR (400 MHz, CDCl 3) delta 10.23 (br s, 1H), 8.55 (s, 1H), 6.66 (t, J=8.0 Hz, 2H), 4.82-4.61 (m, 3H), 4.42-4.36 (m, 2H), 3.37 (s, 2H), 3.22-3.17 (m, 1H), 2.23-2.20 (m, 1H), 1.95-1.89 (m, 1H), 1.77-1.74 (m, 1H), 1.60-1.57 (m, 1H).
Peak 2: MS-ESI (m/z) 423.1[ M+H ] +.1H NMR (400 MHz, CDCl 3) delta 10.25 (br s, 1H), 8.58 (s, 1H), 6.66 (t, J=8.0 Hz, 2H), 4.82-4.61 (m, 3H), 4.44-4.36 (m, 2H), 3.39 (s, 2H), 3.22-3.18 (m, 1H), 2.23-2.20 (m, 1H), 1.95-1.89 (m, 1H), 1.76-1.74 (m, 1H), 1.61-1.57 (m, 1H).
Example 110: HIV MT-4 antiviral and cytotoxicity assays
Antiviral assay in MT-4 cells
Compounds were tested for their ability to inhibit HIV-1 (IIIB) replication in MT-4 cells in a high throughput 384 well assay format. Serial dilutions (1:3) in DMSO were made on 384-well polypropylene plates and further diluted 200-fold using Biotek Micro Flow and labyte ECHO acoustic dispensers to complete RPMI medium (10% fbs,1% p/S). Each plate contained up to 8 test compounds with negative (no drug control) and 5 μm AZT positive control. MT-4 cells were pre-infected with 10. Mu.L of fresh 1:250 dilutions of RPMI (mock infection) or HIV-1IIIB concentrated virus stock. Infected and uninfected MT-4 cells were further diluted in complete RPMI medium and added to each plate using a Micro Flow dispenser. After 5 days of incubation in a humidified and temperature-controlled incubator (37 ℃), cell Titer Glo (Promega) was added to the assay plate and the Envision plate reader was allowed to read for chemiluminescence. EC is to be EC 50 Values are defined as the concentration of the compound that caused a 50% decrease in luminescence signal, and are calculated using an S-type dose response model to generate a curve fit.
Cytotoxicity assays in MT-4 cells
Assays were performed as above except that uninfected MT-4 cells were added to each well containing the test compound. In addition, 10 μm puromycin was added to the last column of each assay plate to assess basal levels of cytotoxicity.
Example 111: HIV MT-4 serum change antiviral reporting assay
To quantify the amount of protein bound to human serum, compounds were serially diluted in DMSO (1:3) and acoustically transferred via labyte ECHO robot to 384 well assay plates. Each plate contained up to 8 test compounds, including negative and positive controls (DMSO, 5 μm AZT, respectively). Assay plates were prepared in duplicate and tested in CCM (cell culture medium) or HS/CCM (human serum/cell culture medium). MT-4 cells were first pre-infected with pLai RLuc reporter virus at 37℃for 2 hours, then further diluted in CCM (RPMI medium, 10% FBS,1% P/S) or HS/CCM (RPMI medium, 10% FBS,50% HS,1% P/S) and then added to each plate using a Biotek Micro Flow dispenser. After 72 hours of incubation in a humidified and temperature-controlled incubator (37 ℃), renilla Glo (Promega) was added to all assay plates and the Envision plate reader was allowed to read for chemiluminescence. EC is to be EC 50 Values are defined as the concentration of the compound that caused a 50% decrease in luminescence signal, and are calculated using an S-type dose response model to generate a curve fit. To determine the amount of protein binding, the amount of protein bound was determined by combining EC 50 (HS/CCM)/EC 50 Dividing (CCM) by calculating EC 50 Fold change (or EC) 50 Change).
Compounds of the present disclosure show antiviral activity in this assay, as shown in table 1 below. Thus, the compounds of the embodiments disclosed herein are useful for treating proliferation of the HIV virus, treating AIDS, or delaying the onset of AIDS or ARC symptoms.
TABLE 1
/>
/>
The data in table 1 represent the average over time for each assay for each compound. For certain compounds, various assays have been performed.
Example 112: half-life of exemplary Compounds
Non-clinical in vivo studies
All non-clinical studies were conducted in accordance with the guidelines for laboratory animal care and use, adopted and published by the national institutes of health, and approved by the institutional animal care and use committee or local equivalent.
Test compound PK was determined in monkeys (Labcorp, madison, WI) 30 minutes after single dose intravenous infusion. For intravenous administration in monkeys, the dosing vehicle was 10% n-methylpyrrolidone; 50% polyethylene glycol 300 and 40% water (pH 8.3). Serial blood samples were collected at 0.25, 0.48 (before the end of infusion), 0.58, 0.75, 1, 1.5, 2, 4, 8, 12, 24, 48, 72, 96, 120, 144, and 168 hours (based on the start of infusion) prior to dosing, and plasma test compound concentrations were quantified using LC-MS/MS methods.
Plasma bioassay
Non-clinical plasma samples were prepared by adding 200 μl cold acetonitrile/internal standard solution to 96-well plates containing 50 μl aliquots of each plasma sample. After protein precipitation, each supernatant (50 μl) was transferred to a clean 96-well plate and diluted with 450 μl water. The sample injection volume was 2 μl. Samples were analyzed by the multiplex reaction monitoring LC-MS/MS method. The LC-MS system consisted of a Cohesive LX-2 multiplexing with two identical DIONEX UltiMate 3000RS pumps, a Hypersil Gold C18 HPLC column (50X 2.1mm,1.9 μm particle size; thermo Fisher Scientific) and an ABSciex QTRAP5500 mass spectrometer LC-MS/MS system. Chromatography was performed using 0.1% formic acid and 1% isopropanol (MP a) in aqueous solution and 0.1% formic acid and 1% isopropanol (MP B) in acetonitrile. The flow rate was maintained at 0.5mL/min (gradient: 0-0.5 min, 5% MP B;1.83 min, 30% MP B;2.83-3.50 min, 95% MP B;3.5-5 min, 5% MP B).
Pharmacokinetic analysis
Plasma PK parameters of test compounds in monkeys were estimated by non-compartmental analysis using software program Phoenix WinNonlin version 6.3 (Pharsight Corporation, mountain View, CA). The following plasma PK parameters (as the case may be) were estimated in non-clinical species: the peak plasma concentration observed (C max ) Reach C max Time (T) max ) Finally, the plasma concentration (C) Finally )、t 1/2 、C Finally Time (T) Finally ) From time 0 to C Finally Area under the plasma concentration-time curve (AUC) Finally ) AUC (AUC) from time 0 to infinity inf ) CL, mean Residence Time (MRT) and steady state distribution volume (V) ss )。
Abbreviations (abbreviations)
AUC: area under plasma concentration-time curve;
AUCinf: area under the plasma concentration time curve from time 0 to infinity;
CL: the clearance rate;
cmax is as follows: the peak plasma concentration observed;
HPLC: high performance liquid chromatography;
LC-MS/MS: liquid chromatography-tandem mass spectrometry;
PK: pharmacokinetics;
tmax: the time to peak plasma concentration;
vss: a steady state distribution volume;
/>
/>
to the extent not inconsistent with the present specification, all U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications, and non-patent publications mentioned in the present specification are incorporated herein by reference in their entirety.
From the foregoing, it will be appreciated that, although specific embodiments have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the disclosure. Accordingly, the disclosure is not limited except as by the appended claims.

Claims (135)

1. A compound of formula I:
or a pharmaceutically acceptable salt thereof, wherein
Ar is C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S; wherein said C 6 -C 10 Aryl or six to ten membered heteroaryl optionally being 1 to 4 independently selected from halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl and C 1 -C 6 Substituent substitution of the group consisting of alkoxy;
R 1 h, C of a shape of H, C 1 -C 3 Alkyl or phenyl;
R 2 is H or C 1 -C 3 An alkyl group;
R 3 is H or C 1 -C 3 An alkyl group;
R 4 and R is 5 Each independently is H, halogen, cyano, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or containing one, two or three members selected from N, O and SSix to ten membered heteroaryl groups of heteroatoms; wherein said C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or six-to ten-membered heteroaryl optionally being independently selected from halogen, C 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; or alternatively
R 4 And R is 5 Taken together form a 3-6 membered carbocyclic ring or a 4-6 membered heterocyclic ring containing one heteroatom selected from N, O and S;
R 6 is H, halogen or C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group;
R 7 is H, halogen or C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group;
R 8A and R is 8B H, C independently 1 -C 3 Alkyl or benzyl; and is also provided with
-X-Y-is- (CR) 13A R 13B ) p -CR 9 =CR 10 -, -or- (CR) 13A R 13B ) q -CR 11A R 11B -CR 12A R 12B -; wherein the method comprises the steps of
R 9 Is H, halogen or C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or C 1 -C 6 An alkoxy group;
R 10 is H, halogen or C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or C 1 -C 6 An alkoxy group; or alternatively
R 9 And R is 10 Together with the carbon to which they are attached, form a phenyl group or a 5-6 membered heteroaryl ring containing 1, 2 or 3 heteroatoms independently selected from N, O and S; wherein said phenyl or said 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 Substituent substitution of the group consisting of alkoxy; and is also provided with
R 11A 、R 11B 、R 12A 、R 12B 、R 13A And R is 13B Each independently is H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 An alkoxy group; or C 1 -C 6 A haloalkyl group; or alternatively
R 11A 、R 12A 、R 13A And R is 13B Each independently is H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group; and R is 11B And R is 12B Together with the carbon to which they are attached, form a 3-6 membered carbocyclic ring; wherein the 3-6 membered carbocycle is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 Substituent substitution of the group consisting of alkoxy;
p is 0 or 1;
q is 0 or 1;
wherein when-X-Y is- (CR) 13A R 13B ) q -CR 11A R 11B -CR 12A R 12B -when, then:
(v)R 4 is halogen, cyano, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S; wherein said C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or six-to ten-membered heteroaryl optionally being independently selected from halogen, C 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; and R is 5 Is H, halogen, cyano, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S; wherein said C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or six-to ten-membered heteroaryl optionally being independently selected from halogen, C 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; or alternatively
(vi)R 4 And R is 5 Are linked together to form a 3-6 membered carbocyclic ring or a 4-6 membered heterocyclic ring having one heteroatom; or alternatively
(vii)R 8A Is C 1 -C 3 Alkyl or benzyl; or alternatively
(viii)R 6 Is halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound has formula Ia:
3. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound has formula Ib:
4. a compound according to any one of claims 1-3, or a pharmaceutically acceptable salt thereof, wherein Ar is C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one heteroatom selected from N, O and S; wherein said C 6 -C 10 Aryl or six to ten membered heteroaryl optionally being 1 to 4 independently selected from halogen, C 1 -C 6 Alkyl and C 1 -C 6 The substituents of the group consisting of alkoxy groups.
5. According to claim1-4, or a pharmaceutically acceptable salt thereof, wherein Ar is C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one heteroatom selected from N, O and S; wherein said C 6 -C 10 Aryl or six to ten membered heteroaryl groups are optionally substituted with 1-4 halogens.
6. The compound according to any one of claims 1-5, or a pharmaceutically acceptable salt thereof, wherein Ar is C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one heteroatom selected from N, O and S; wherein said C 6 -C 10 Aryl or six to ten membered heteroaryl is optionally substituted with 1-3 substituents independently selected from Cl and F.
7. The compound of any one of claims 1-6, or a pharmaceutically acceptable salt thereof, wherein Ar is a group of the formula:
wherein Z is N or CR A The method comprises the steps of carrying out a first treatment on the surface of the n is 0, 1, 2, 3 or 4; and each R A Independently is halogen, C 1 -C 6 Alkyl and C 1 -C 6 An alkoxy group.
8. The compound of claim 7, or a pharmaceutically acceptable salt thereof, wherein Z is CH or N.
9. The compound according to claim 7 or 8, or a pharmaceutically acceptable salt thereof, wherein Z is CH.
10. The compound according to claim 7 or 8, or a pharmaceutically acceptable salt thereof, wherein Z is N.
11. The compound according to any one of claims 1-9, or a pharmaceutically acceptable salt thereof, wherein Ar isn is 1, 2, 3 or 4; and each R A Independently selected from halogen and C 1 -C 6 An alkoxy group.
12. A compound according to any one of claims 1-9 and 11, or a pharmaceutically acceptable salt thereof, ar isn is 1, 2, 3 or 4; and each R A Independently halo or-O-C 1-4 An alkyl group.
13. The compound according to any one of claims 1-9, 11 and 12, or a pharmaceutically acceptable salt thereof, wherein Ar is
14. The compound according to any one of claims 1-8 and 10, or a pharmaceutically acceptable salt thereof, wherein Ar isn is 1, 2 or 3, and each R A Independently fluorine or chlorine.
15. The compound of any one of claims 1-9 and 11-13, or a pharmaceutically acceptable salt thereof, wherein the compound has formula II:
wherein the method comprises the steps of
n is 0, 1, 2, 3 or 4; and is also provided with
Each R A Independently is halogen, C 1 -C 6 Alkyl and C 1 -C 6 An alkoxy group.
16. The compound according to any one of claims 1-9, 11-13 and 15, or a pharmaceutically acceptable salt thereof, wherein the compound has formula IIa:
Wherein the method comprises the steps of
n is 0, 1, 2, 3 or 4; and is also provided with
Each R A Independently is halogen, C 1 -C 6 Alkyl and C 1 -C 6 An alkoxy group.
17. The compound of any one of claims 1-9, 11-13, and 15, or a pharmaceutically acceptable salt thereof, wherein the compound has formula IIb:
wherein the method comprises the steps of
n is 0, 1, 2, 3 or 4; and is also provided with
Each R A Independently is halogen, C 1 -C 6 Alkyl and C 1 -C 6 An alkoxy group.
18. A compound according to any one of claims 1-17, or a pharmaceutically acceptable salt thereof, wherein-X-Y-is- (CR) 13A R 13B ) q -CR 9 =CR 10 -。
19. The compound of any one of claims 1-18, or a pharmaceutically acceptable salt thereof, wherein-X-Y-is-CR 13A R 13B -CR 9 =CR 10 -。
20. The method according to any one of claims 1-19The compound or a pharmaceutically acceptable salt thereof, wherein-X-Y-is-CH 2 -CR 9 =CR 10 -。
21. The compound of any one of claims 1-18, or a pharmaceutically acceptable salt thereof, wherein-X-Y-is-CR 9 =CR 10 -。
22. The compound of any one of claims 1-21, or a pharmaceutically acceptable salt thereof, wherein R 9 And R is 10 Together with the carbon to which they are attached, form a phenyl group or a 5-6 membered heteroaryl ring containing 1, 2 or 3 heteroatoms independently selected from N, O and S; wherein said phenyl or said 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
23. The compound of any one of claims 1-22, or a pharmaceutically acceptable salt thereof, wherein R 9 And R is 10 Together with the carbon to which they are attached, form a phenyl group or a 5-6 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms independently selected from N and O; wherein said phenyl or said 5-6 membered heteroaromatic ring is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
24. The compound of any one of claims 1-23, or a pharmaceutically acceptable salt thereof, wherein R 9 Is H or halogen; and R is 10 Is H or halogen; or R is 9 And R is 10 Together with the carbon to which they are attached, form a phenyl group, wherein the phenyl group is optionally substituted with 1, 2 or 3 groups independently selected from halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
25. The compound of any one of claims 1-24, or a pharmaceutically acceptable salt thereof, wherein R 9 Is H or halogen; and R is 10 Is H or halogen; or R is 9 And R is 10 Together with the carbon to which they are attached, form a phenyl group, wherein the phenyl group is optionally substituted with one member selected from the group consisting of halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl and C 1 -C 3 The substituents of the group consisting of alkoxy groups.
26. The compound of any one of claims 1-25, or a pharmaceutically acceptable salt thereof, wherein R 9 Is H or halogen; and R is 10 Is H or halogen; or R is 9 And R is 10 Together with the carbon to which they are attached, form a phenyl group, wherein the phenyl group is optionally substituted with one halogen.
27. The compound of any one of claims 1 to 26, or a pharmaceutically acceptable salt thereof, wherein the compound has formula III:
wherein the method comprises the steps of
m is 0, 1, 2 or 3; and is also provided with
Each R B Independently is halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or C 1 -C 3 An alkoxy group.
28. The compound of any one of claims 1-26 and 27, or a pharmaceutically acceptable salt thereof, wherein the compound has formula IIIa:
wherein the method comprises the steps of
m is 0, 1, 2 or 3; and is also provided with
Each R B Independently is halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or C 1 -C 3 An alkoxy group.
29. The compound of any one of claims 1-26 and 27, or a pharmaceutically acceptable salt thereof, wherein the compound has formula IIIb:
wherein the method comprises the steps of
m is 0, 1, 2 or 3; and is also provided with
Each R B Independently is halogen, C 1 -C 3 Alkyl, C 1 -C 3 Haloalkyl or C 1 -C 3 An alkoxy group.
30. The compound of any one of claims 1 to 26, or a pharmaceutically acceptable salt thereof, wherein the compound has formula IV:
Wherein z is 0 or 1.
31. The compound of any one of claims 1-26 and 30, or a pharmaceutically acceptable salt thereof, wherein the compound has formula IVa:
wherein z is 0 or 1.
32. The compound of any one of claims 1-26 and 30, or a pharmaceutically acceptable salt thereof, wherein the compound has formula IVb:
wherein z is 0 or 1.
33. The compound of any one of claims 1-32, or a pharmaceutically acceptable salt thereof, wherein each R 9 And R is 10 Independently H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Haloalkyl or C 1 -C 6 An alkoxy group.
34. The compound of any one of claims 1-33, or a pharmaceutically acceptable salt thereof, wherein each R 9 And R is 10 Independently H, halogen, C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group.
35. The compound of any one of claims 1-34, or a pharmaceutically acceptable salt thereof, wherein each R 9 And R is 10 Independently H or halogen.
36. The compound according to any one of claims 29 to 35, or a pharmaceutically acceptable salt thereof, wherein z is 0.
37. A compound according to any one of claims 29 to 35, or a pharmaceutically acceptable salt thereof, wherein z is 1.
38. The compound of claim 30, or a pharmaceutically acceptable salt thereof, wherein:
z is 0;
R 1 、R 2 and R is 3 Each is H;
R 4 and R is 5 Independently H, halogen, cyano, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 6 -C 10 Aryl or a six to ten membered heteroaryl containing one, two or three heteroatoms selected from N, O and S;
R 6 and R is 7 Independently H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group;
R 8A and R is 8B All are H;
R 9 and R is 10 Each independently is H, halogen, C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group;
each R A Independently fluorine or chlorine;
n is 1, 2 or 3.
39. The compound of claim 30, or a pharmaceutically acceptable salt thereof, wherein:
z is 0;
R 1 、R 2 and R is 3 Each is H;
R 4 and R is 5 Independently H, halogen, cyano, C 1 -C 6 Alkyl or C 1 -C 6 An alkoxy group;
R 6 and R is 7 Independently H, halogen, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy or C 1 -C 6 A haloalkyl group;
R 8A and R is 8B All are H;
R 9 and R is 10 Each independently is H, halogen, C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group;
each R A Independently fluorine or chlorine;
n is 1, 2 or 3.
40. The compound of claim 30, or a pharmaceutically acceptable salt thereof, wherein:
z is 0;
R 1 、R 2 and R is 3 Each is H;
R 4 and R is 5 Independently H, halogen or C 1 -C 6 An alkyl group;
R 6 and R is 7 Independently H, halogen or C 1 -C 6 An alkyl group;
R 8A and R is 8B All are H;
R 9 and R is 10 Each independently is H, halogen or C 1 -C 6 An alkyl group;
Each R A Independently fluorine or chlorine;
n is 1, 2 or 3.
41. A compound according to any one of claims 1-17, or a pharmaceutically acceptable salt thereof, wherein-X-Y-is- (CR) 13A R 13B ) q -CR 11A R 11B -CR 12A R 12B -。
42. A compound according to one of claims 1-17 and 41, or a pharmaceutically acceptable salt thereof, wherein-X-Y-is-CR 13A R 13B -CR 11A R 11B -CR 12A R 12B -。
43. A compound according to any one of claims 1-17 and 41, or a pharmaceutically acceptable salt thereof, wherein-X-Y-is-CR 11A R 11B -CR 12A R 12B -。
44. The compound of any one of claims 1-17 and 43, or a pharmaceutically acceptable salt thereof, wherein the compound has formula V:
wherein z' is 1 or 2.
45. The compound of any one of claims 1-17, 43, and 44, or a pharmaceutically acceptable salt thereof, wherein the compound has formula Va:
wherein z' is 1 or 2.
46. The compound of any one of claims 1-17, 43, and 44, or a pharmaceutically acceptable salt thereof, wherein the compound has formula Vb:
wherein z' is 1 or 2.
47. The compound of any one of claims 44-46, or a pharmaceutically acceptable salt thereof, wherein z' is 1.
48. The compound of any one of claims 44-46, or a pharmaceutically acceptable salt thereof, wherein z' is 2.
49. The compound of any one of claims 1-48, or a pharmaceutically acceptable salt thereof, wherein R 8B H.
50. The compound of any one of claims 1-49, or a pharmaceutically acceptable salt thereof, wherein R 8A H.
51. The compound of any one of claims 1-50, or a pharmaceutically acceptable salt thereof, wherein R 3 H.
52. A compound according to any one of claims 1-51 or a pharmaceutically acceptable thereofAcceptable salts, wherein R 1 And R is 2 All are H.
53. The compound of any one of claims 1-52, or a pharmaceutically acceptable salt thereof, wherein each R 4 And R is 5 Independently H or C 1 -C 6 Alkyl, wherein the C 1 -C 6 Alkyl groups optionally being independently selected from halogen, C by one, two or three 1 -C 3 Alkoxy or C 1 -C 3 The group substitution of haloalkoxy groups.
54. The compound of any one of claims 1-53, or a pharmaceutically acceptable salt thereof, wherein R 4 Is H or C 1 -C 6 Alkyl, wherein the C 1 -C 6 Alkyl groups optionally being independently selected from halogen, C by one, two or three 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; and R is 5 Is H or C 1 -C 6 An alkyl group.
55. The compound of any one of claims 1-54, or a pharmaceutically acceptable salt thereof, wherein R 4 Is H or C 1 -C 6 Alkyl, wherein the C 1 -C 6 Alkyl groups optionally being independently selected from halogen, C by one, two or three 1 -C 3 Alkoxy or C 1 -C 3 Substitution of the groups of the haloalkoxy groups; and R is 5 H.
56. The compound of any one of claims 1-52, or a pharmaceutically acceptable salt thereof, wherein R 4 And R is 5 Independently H, me, OMe or-CH 2 F。
57. The compound of any one of claims 1-52, or a pharmaceutically acceptable salt thereof, wherein R 4 And R is 5 Are linked together to form a 4-6 membered heterocyclic ring containing one heteroatom selected from N, O and S.
58. The compound of any one of claims 1-57, or a pharmaceutically acceptable salt thereof, wherein R 6 Is H, halogen or C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group.
59. The compound of any one of claims 1-55, or a pharmaceutically acceptable salt thereof, wherein R 6 H, C of a shape of H, C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group.
60. The compound of any one of claims 1-59, or a pharmaceutically acceptable salt thereof, wherein R 6 Is H, methyl or CH 2 F。
61. The compound of any one of claims 1-58, or a pharmaceutically acceptable salt thereof, wherein R 6 Is H, halogen or C 1 -C 6 An alkyl group.
62. The compound of any one of claims 1-61, or a pharmaceutically acceptable salt thereof, wherein R 6 Is halogen or C 1 -C 6 An alkyl group.
63. The compound of any one of claims 1-62, or a pharmaceutically acceptable salt thereof, wherein R 6 Is C 1 -C 6 An alkyl group.
64. The compound of any one of claims 1-63, or a pharmaceutically acceptable salt thereof, wherein R 6 Is methyl.
65. The compound of any one of claims 1-61, or a pharmaceutically acceptable salt thereof, wherein R 6 H.
66. The chemical process of any one of claims 1-65A compound or pharmaceutically acceptable salt thereof, wherein R 7 Is H, halogen or C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group.
67. The compound of any one of claims 1-66, or a pharmaceutically acceptable salt thereof, wherein R 7 Is H, halogen or C 1 -C 6 An alkyl group.
68. The compound of any one of claims 1-67, or a pharmaceutically acceptable salt thereof, wherein R 7 Is halogen or C 1 -C 6 An alkyl group.
69. The compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, wherein R 7 Is C 1 -C 6 An alkyl group.
70. The compound of any one of claims 1-67, or a pharmaceutically acceptable salt thereof, wherein R 7 H.
71. The compound of any one of claims 1-70, or a pharmaceutically acceptable salt thereof, wherein R 11A 、R 11B 、R 12A 、R 12B 、R 13A And R is 13B Each independently is H, halogen, C 1 -C 6 Alkyl or C 1 -C 6 An alkoxy group.
72. The compound of any one of claims 1-71, or a pharmaceutically acceptable salt thereof, wherein R 11A 、R 11B 、R 12A 、R 12B 、R 13A And R is 13B Each independently is H, halo or C 1 -C 6 An alkoxy group.
73. The compound of any one of claims 1-72, or a pharmaceutically acceptable salt thereof, wherein R 11A 、R 11B 、R 12A 、R 12B 、R 13A And R is 13B Each independently is H, halogen or methoxy.
74. The compound of any one of claims 1-73, or a pharmaceutically acceptable salt thereof, wherein R 11A 、R 11B 、R 12A 、R 12B 、R 13A And R is 13B Each independently is H, fluoro or methoxy.
75. A compound having the formula:
or a pharmaceutically acceptable salt thereof.
76. A compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
/>
77. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
/>
/>
/>
/>
78. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
/>
/>
/>
79. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
/>
/>
80. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
/>
/>
81. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
82. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is:
83. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is:
84. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is:
85. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is:
86. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is:
87. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is:
88. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is:
89. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is:
/>
90. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is:
91. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is:
92. the compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is:
93. a compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of:
/>
/>
94. a pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of claims 1-93, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
95. The pharmaceutical composition of claim 94, further comprising an additional therapeutic agent.
96. The pharmaceutical composition of claim 95, wherein the additional therapeutic agent is an anti-HIV agent.
97. The pharmaceutical composition of claim 95 or 96, wherein the additional therapeutic agent is an HIV protease inhibitor, a non-nucleoside or non-nucleotide inhibitor of HIV reverse transcriptase, a nucleoside or nucleotide inhibitor of HIV reverse transcriptase, an HIV capsid inhibitor, a gp41 inhibitor, a CXCR4 inhibitor, a gp120 inhibitor, a CCR5 inhibitor, a latency reversing agent, a capsid polymerization inhibitor, an HIV bNAb, a TLR7 agonist, a pharmacokinetic enhancer, or a combination thereof.
98. The pharmaceutical composition of any one of claims 95-97, wherein said additional therapeutic agent is lenacipam, isala Qu Wei, or a pharmaceutically acceptable salt thereof.
99. The pharmaceutical composition of any one of claims 95-97, wherein said additional therapeutic agent is abacavir, tenofovir alafenamide, tenofovir disoproxil, lenacipam, GS-5894, isala Qu Wei, or a pharmaceutically acceptable salt thereof.
100. The pharmaceutical composition of any one of claims 95-99, wherein the pharmaceutical composition is for oral or parenteral administration.
101. A kit comprising a compound of any one of claims 1-93, or a pharmaceutically acceptable salt thereof, and instructions for use.
102. The kit of claim 101, further comprising an additional therapeutic agent.
103. The kit of claim 102, wherein the additional therapeutic agent is an anti-HIV agent.
104. The kit of claim 102 or 103, wherein the additional therapeutic agent is an HIV protease inhibitor, a non-nucleoside or non-nucleotide inhibitor of HIV reverse transcriptase, a nucleoside or nucleotide inhibitor of HIV reverse transcriptase, an HIV capsid inhibitor, a gp41 inhibitor, a CXCR4 inhibitor, a gp120 inhibitor, a CCR5 inhibitor, a latency reversing agent, a capsid polymerization inhibitor, HIV bNAb, a TLR7 agonist, a pharmacokinetic enhancer, or a combination thereof.
105. The kit of any one of claims 102-104, wherein the additional therapeutic agent is lenacipam, isala Qu Wei, or a pharmaceutically acceptable salt thereof.
106. The kit of any one of claims 102-104, wherein the one or more additional therapeutic agents is abacavir, tenofovir alafenamide, tenofovir disoproxil, lenacipam, GS-5894, isala Qu Wei, or a pharmaceutically acceptable salt thereof.
107. A method of treating HIV infection in a human suffering from or at risk of suffering from HIV infection, the method comprising administering to the human a therapeutically effective amount of a compound according to any one of claims 1-93, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to any one of claims 94-100.
108. The method of claim 107, further comprising administering to the human a therapeutically effective amount of an additional therapeutic agent.
109. The method of claim 108, wherein the additional therapeutic agent is an anti-HIV agent.
110. The method of claim 108 or 109, wherein the additional therapeutic agent is an HIV protease inhibitor, a non-nucleoside or non-nucleotide inhibitor of HIV reverse transcriptase, a nucleoside or nucleotide inhibitor of HIV reverse transcriptase, an HIV capsid inhibitor, a gp41 inhibitor, a CXCR4 inhibitor, a gp120 inhibitor, a CCR5 inhibitor, a latency reversing agent, a capsid polymerization inhibitor, HIV bNAb, a TLR7 agonist, a pharmacokinetic enhancer, or a combination thereof.
111. The method of any one of claims 108-110 wherein the additional therapeutic agent is lenacipam, isala Qu Wei, or a pharmaceutically acceptable salt thereof.
112. The method of any one of claims 108-110, wherein the one or more additional therapeutic agents is abacavir, tenofovir alafenamide, tenofovir disoproxil, lenacipam, GS-5894, isala Qu Wei, or a pharmaceutically acceptable salt thereof.
113. The method of any one of claims 107-112, wherein the administration is oral, intravenous, subcutaneous, or intramuscular.
114. Use of a compound according to any one of claims 1-93, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to any one of claims 94-100, for treating HIV infection in a human suffering from or at risk of suffering from HIV infection.
115. The use of claim 114, wherein the compound is used with a therapeutically effective amount of an additional therapeutic agent.
116. The use of claim 115, wherein the additional therapeutic agent is an anti-HIV agent.
117. The use of claim 115 or 116, wherein the additional therapeutic agent is an HIV protease inhibitor, a non-nucleoside or non-nucleotide inhibitor of HIV reverse transcriptase, a nucleoside or nucleotide inhibitor of HIV reverse transcriptase, an HIV capsid inhibitor, a gp41 inhibitor, a CXCR4 inhibitor, a gp120 inhibitor, a CCR5 inhibitor, a latency reversing agent, a capsid polymerization inhibitor, HIV bNAb, a TLR7 agonist, a pharmacokinetic enhancer, or a combination thereof.
118. The use of any one of claims 115-117, wherein the additional therapeutic agent is lenacipam, isala Qu Wei, or a pharmaceutically acceptable salt thereof.
119. The use of any one of claims 115-117, wherein the one or more additional therapeutic agents is abacavir, tenofovir alafenamide, tenofovir disoproxil, lenacipam, GS-5894, isala Qu Wei, or a pharmaceutically acceptable salt thereof.
120. The use of any one of claims 115-119, wherein the compound is used by oral, intravenous, subcutaneous, or intramuscular administration.
121. A compound according to any one of claims 1-93, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to any one of claims 94-100, for use in medical therapy.
122. A compound according to any one of claims 1-93, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to any one of claims 94-100, for use in the treatment of HIV infection.
123. The compound of claim 122, wherein the compound is used with a therapeutically effective amount of an additional therapeutic agent.
124. The compound of claim 123, wherein the additional therapeutic agent is an anti-HIV agent.
125. The compound of claim 123 or 124, wherein the additional therapeutic agent is an HIV protease inhibitor, a non-nucleoside or non-nucleotide inhibitor of HIV reverse transcriptase, a nucleoside or nucleotide inhibitor of HIV reverse transcriptase, an HIV capsid inhibitor, a gp41 inhibitor, a CXCR4 inhibitor, a gp120 inhibitor, a CCR5 inhibitor, a latency reversing agent, a capsid polymerization inhibitor, HIV bNAb, a TLR7 agonist, a pharmacokinetic enhancer, or a combination thereof.
126. The compound of any one of claims 123-125, wherein the additional therapeutic agent is lenacipam, isala Qu Wei, or a pharmaceutically acceptable salt thereof.
127. The compound of any one of claims 123-125, wherein the one or more additional therapeutic agents is abacavir, tenofovir alafenamide, tenofovir disoproxil, lenacipam, GS-5894, isala Qu Wei, or a pharmaceutically acceptable salt thereof.
128. The compound of any of claims 122-127, wherein the compound is for use by oral, intravenous, subcutaneous, or intramuscular administration.
129. Use of a compound according to any one of claims 1-93, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to any one of claims 94-100, in the manufacture of a medicament for treating HIV infection in a human suffering from or at risk of suffering from HIV infection.
130. The use of claim 129, wherein the medicament is for use with a therapeutically effective amount of an additional therapeutic agent.
131. The use of claim 130, wherein the additional therapeutic agent is an anti-HIV agent.
132. The use of claim 130 or 131, wherein the additional therapeutic agent is an HIV protease inhibitor, a non-nucleoside or non-nucleotide inhibitor of HIV reverse transcriptase, a nucleoside or nucleotide inhibitor of HIV reverse transcriptase, an HIV capsid inhibitor, a gp41 inhibitor, a CXCR4 inhibitor, a gp120 inhibitor, a CCR5 inhibitor, a latency reversing agent, a capsid polymerization inhibitor, HIV bNAb, a TLR7 agonist, a pharmacokinetic enhancer, or a combination thereof.
133. The use of any one of claims 130-132, wherein the additional therapeutic agent is lenacipam, isala Qu Wei, or a pharmaceutically acceptable salt thereof.
134. The use of any one of claims 130-132, wherein the one or more additional therapeutic agents is abacavir, tenofovir alafenamide, tenofovir disoproxil, lenacipam, GS-5894, isala Qu Wei, or a pharmaceutically acceptable salt thereof.
135. The use of any of claims 129-134, wherein the compound is used by oral, intravenous, subcutaneous, or intramuscular administration.
CN202280010649.4A 2021-01-19 2022-01-18 Substituted pyridotriazine compounds and uses thereof Pending CN116745292A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/139,237 2021-01-19
US202163190461P 2021-05-19 2021-05-19
US63/190,461 2021-05-19
PCT/US2022/012773 WO2022159387A1 (en) 2021-01-19 2022-01-18 Substituted pyridotriazine compounds and uses thereof

Publications (1)

Publication Number Publication Date
CN116745292A true CN116745292A (en) 2023-09-12

Family

ID=87910146

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280010649.4A Pending CN116745292A (en) 2021-01-19 2022-01-18 Substituted pyridotriazine compounds and uses thereof

Country Status (1)

Country Link
CN (1) CN116745292A (en)

Similar Documents

Publication Publication Date Title
CN110536889B (en) HIV inhibitor compounds
US10646486B2 (en) Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
TWI829205B (en) Anti-hiv compounds
CN116390924A (en) Bridged tricyclic carbamoyl pyridone compounds and uses thereof
US11897892B2 (en) Substituted pyridotriazine compounds and uses thereof
CN115151545A (en) Tetracyclic compounds for the treatment of HIV infection
CN116745292A (en) Substituted pyridotriazine compounds and uses thereof
TW202408533A (en) Substituted pyridotriazine compounds and uses thereof
KR20220145368A (en) Tetracyclic compounds for the treatment of HIV infection
EA040208B1 (en) ANALOGUES OF ATAZANAVIR (ATV) FOR THE TREATMENT OF HIV INFECTIONS

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40094825

Country of ref document: HK